WO1993004191A1 - Conjugues de toxines non cytolytiques - Google Patents
Conjugues de toxines non cytolytiques Download PDFInfo
- Publication number
- WO1993004191A1 WO1993004191A1 PCT/US1992/006823 US9206823W WO9304191A1 WO 1993004191 A1 WO1993004191 A1 WO 1993004191A1 US 9206823 W US9206823 W US 9206823W WO 9304191 A1 WO9304191 A1 WO 9304191A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- toxin
- cytolytic
- target cell
- moiety
- cytolytic toxin
- Prior art date
Links
- 239000002596 immunotoxin Substances 0.000 title description 16
- 239000003053 toxin Substances 0.000 claims abstract description 246
- 231100000765 toxin Toxicity 0.000 claims abstract description 246
- 230000008685 targeting Effects 0.000 claims abstract description 66
- 238000000034 method Methods 0.000 claims abstract description 41
- 239000003446 ligand Substances 0.000 claims abstract description 35
- 230000001225 therapeutic effect Effects 0.000 claims abstract description 17
- 238000001243 protein synthesis Methods 0.000 claims abstract description 14
- 239000000758 substrate Substances 0.000 claims abstract description 14
- 230000014616 translation Effects 0.000 claims abstract description 14
- 102000007469 Actins Human genes 0.000 claims abstract description 13
- 108010085238 Actins Proteins 0.000 claims abstract description 13
- 230000007727 signaling mechanism Effects 0.000 claims abstract description 12
- 230000008569 process Effects 0.000 claims abstract description 11
- 108700012359 toxins Proteins 0.000 claims description 247
- 210000004027 cell Anatomy 0.000 claims description 183
- 230000001461 cytolytic effect Effects 0.000 claims description 93
- 230000027455 binding Effects 0.000 claims description 68
- 230000002255 enzymatic effect Effects 0.000 claims description 54
- 102000005962 receptors Human genes 0.000 claims description 54
- 108020003175 receptors Proteins 0.000 claims description 54
- 108091006027 G proteins Proteins 0.000 claims description 52
- 102000030782 GTP binding Human genes 0.000 claims description 52
- 108091000058 GTP-Binding Proteins 0.000 claims description 52
- 230000000694 effects Effects 0.000 claims description 52
- 210000004881 tumor cell Anatomy 0.000 claims description 42
- 230000035755 proliferation Effects 0.000 claims description 38
- 230000008512 biological response Effects 0.000 claims description 36
- 108010049048 Cholera Toxin Proteins 0.000 claims description 33
- 102000009016 Cholera Toxin Human genes 0.000 claims description 32
- 229930186900 holotoxin Natural products 0.000 claims description 32
- 239000003795 chemical substances by application Substances 0.000 claims description 29
- 206010028980 Neoplasm Diseases 0.000 claims description 28
- 239000012636 effector Substances 0.000 claims description 27
- 210000001744 T-lymphocyte Anatomy 0.000 claims description 26
- 108090000765 processed proteins & peptides Proteins 0.000 claims description 26
- 230000005847 immunogenicity Effects 0.000 claims description 23
- 230000005764 inhibitory process Effects 0.000 claims description 20
- 108010081690 Pertussis Toxin Proteins 0.000 claims description 19
- 230000005730 ADP ribosylation Effects 0.000 claims description 17
- 108060000200 adenylate cyclase Proteins 0.000 claims description 16
- 102000030621 adenylate cyclase Human genes 0.000 claims description 16
- 102000036639 antigens Human genes 0.000 claims description 14
- 108091007433 antigens Proteins 0.000 claims description 14
- 238000011282 treatment Methods 0.000 claims description 14
- 102000004127 Cytokines Human genes 0.000 claims description 13
- 108090000695 Cytokines Proteins 0.000 claims description 13
- 108010002350 Interleukin-2 Proteins 0.000 claims description 13
- 102000000588 Interleukin-2 Human genes 0.000 claims description 13
- 239000000427 antigen Substances 0.000 claims description 13
- 238000001727 in vivo Methods 0.000 claims description 12
- 230000007246 mechanism Effects 0.000 claims description 12
- 230000001105 regulatory effect Effects 0.000 claims description 12
- 108091005804 Peptidases Proteins 0.000 claims description 11
- 239000004365 Protease Substances 0.000 claims description 11
- 230000001965 increasing effect Effects 0.000 claims description 11
- 230000001404 mediated effect Effects 0.000 claims description 11
- 230000002503 metabolic effect Effects 0.000 claims description 11
- 102000004196 processed proteins & peptides Human genes 0.000 claims description 10
- 108010083528 Adenylate Cyclase Toxin Proteins 0.000 claims description 9
- 230000036755 cellular response Effects 0.000 claims description 9
- 238000000338 in vitro Methods 0.000 claims description 9
- 210000000130 stem cell Anatomy 0.000 claims description 9
- 241000588832 Bordetella pertussis Species 0.000 claims description 8
- 102000004190 Enzymes Human genes 0.000 claims description 8
- 108090000790 Enzymes Proteins 0.000 claims description 8
- 230000001580 bacterial effect Effects 0.000 claims description 8
- 230000005754 cellular signaling Effects 0.000 claims description 8
- 210000001163 endosome Anatomy 0.000 claims description 8
- 230000002401 inhibitory effect Effects 0.000 claims description 8
- 230000011664 signaling Effects 0.000 claims description 8
- 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 8
- 102100037486 Reverse transcriptase/ribonuclease H Human genes 0.000 claims description 7
- 230000001939 inductive effect Effects 0.000 claims description 7
- 101800003838 Epidermal growth factor Proteins 0.000 claims description 6
- 102400001368 Epidermal growth factor Human genes 0.000 claims description 6
- 102100039620 Granulocyte-macrophage colony-stimulating factor Human genes 0.000 claims description 6
- 108090000901 Transferrin Proteins 0.000 claims description 6
- 102000004338 Transferrin Human genes 0.000 claims description 6
- 210000003719 b-lymphocyte Anatomy 0.000 claims description 6
- 230000004663 cell proliferation Effects 0.000 claims description 6
- 230000007423 decrease Effects 0.000 claims description 6
- 239000000147 enterotoxin Substances 0.000 claims description 6
- 231100000655 enterotoxin Toxicity 0.000 claims description 6
- 229940116977 epidermal growth factor Drugs 0.000 claims description 6
- 239000012581 transferrin Substances 0.000 claims description 6
- 102000008130 Cyclic AMP-Dependent Protein Kinases Human genes 0.000 claims description 5
- 108010049894 Cyclic AMP-Dependent Protein Kinases Proteins 0.000 claims description 5
- 102100036519 Gastrin-releasing peptide Human genes 0.000 claims description 5
- 230000002708 enhancing effect Effects 0.000 claims description 5
- 210000000056 organ Anatomy 0.000 claims description 5
- 238000002560 therapeutic procedure Methods 0.000 claims description 5
- 108010051479 Bombesin Proteins 0.000 claims description 4
- 108010071942 Colony-Stimulating Factors Proteins 0.000 claims description 4
- 101710146739 Enterotoxin Proteins 0.000 claims description 4
- 241000588724 Escherichia coli Species 0.000 claims description 4
- 102000004862 Gastrin releasing peptide Human genes 0.000 claims description 4
- 108090001053 Gastrin releasing peptide Proteins 0.000 claims description 4
- 102000009618 Transforming Growth Factors Human genes 0.000 claims description 4
- 108010009583 Transforming Growth Factors Proteins 0.000 claims description 4
- 230000001363 autoimmune Effects 0.000 claims description 4
- 210000001185 bone marrow Anatomy 0.000 claims description 4
- PUBCCFNQJQKCNC-XKNFJVFFSA-N gastrin-releasingpeptide Chemical compound C([C@@H](C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCSC)C(N)=O)NC(=O)CNC(=O)[C@@H](NC(=O)[C@H](C)NC(=O)[C@H](CC=1C2=CC=CC=C2NC=1)NC(=O)[C@H](CC=1N=CNC=1)NC(=O)[C@H](CC(N)=O)NC(=O)CNC(=O)[C@H](CCCN=C(N)N)NC(=O)[C@H]1N(CCC1)C(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)[C@H](CCSC)NC(=O)[C@H](CCCCN)NC(=O)[C@@H](NC(=O)[C@H](CC(C)C)NC(=O)[C@@H](NC(=O)[C@@H](NC(=O)CNC(=O)CNC(=O)CNC(=O)[C@H](C)NC(=O)[C@H]1N(CCC1)C(=O)[C@H](CC(C)C)NC(=O)[C@H]1N(CCC1)C(=O)[C@@H](N)C(C)C)[C@@H](C)O)C(C)C)[C@@H](C)O)C(C)C)C1=CNC=N1 PUBCCFNQJQKCNC-XKNFJVFFSA-N 0.000 claims description 4
- 239000003102 growth factor Substances 0.000 claims description 4
- 229940088597 hormone Drugs 0.000 claims description 4
- NQDJXKOVJZTUJA-UHFFFAOYSA-N nevirapine Chemical compound C12=NC=CC=C2C(=O)NC=2C(C)=CC=NC=2N1C1CC1 NQDJXKOVJZTUJA-UHFFFAOYSA-N 0.000 claims description 4
- 108030001720 Bontoxilysin Proteins 0.000 claims description 3
- 241001112695 Clostridiales Species 0.000 claims description 3
- BWGNESOTFCXPMA-UHFFFAOYSA-N Dihydrogen disulfide Chemical compound SS BWGNESOTFCXPMA-UHFFFAOYSA-N 0.000 claims description 3
- 208000031886 HIV Infections Diseases 0.000 claims description 3
- 208000037357 HIV infectious disease Diseases 0.000 claims description 3
- 108010002586 Interleukin-7 Proteins 0.000 claims description 3
- 102000004861 Phosphoric Diester Hydrolases Human genes 0.000 claims description 3
- 108090001050 Phosphoric Diester Hydrolases Proteins 0.000 claims description 3
- 231100001103 botulinum neurotoxin Toxicity 0.000 claims description 3
- 230000003247 decreasing effect Effects 0.000 claims description 3
- 239000005556 hormone Substances 0.000 claims description 3
- 208000033519 human immunodeficiency virus infectious disease Diseases 0.000 claims description 3
- 241000193830 Bacillus <bacterium> Species 0.000 claims description 2
- 108060003951 Immunoglobulin Proteins 0.000 claims description 2
- 102000018358 immunoglobulin Human genes 0.000 claims description 2
- 101710160107 Outer membrane protein A Proteins 0.000 claims 2
- 230000035945 sensitivity Effects 0.000 claims 2
- 241000588807 Bordetella Species 0.000 claims 1
- 108090000312 Calcium Channels Proteins 0.000 claims 1
- 102000003922 Calcium Channels Human genes 0.000 claims 1
- 102100021592 Interleukin-7 Human genes 0.000 claims 1
- OKSSKVHGKYJNLL-LJRZAWCWSA-N [(3as,4r,9s,10as)-2,6-diamino-10,10-dihydroxy-9-sulfooxy-3a,4,8,9-tetrahydro-1h-pyrrolo[1,2-c]purin-4-yl]methoxycarbonylsulfamic acid Chemical compound OS(=O)(=O)NC(=O)OC[C@@H]1N=C(N)N2C[C@H](OS(O)(=O)=O)C(O)(O)[C@@]22N=C(N)N[C@H]21 OKSSKVHGKYJNLL-LJRZAWCWSA-N 0.000 claims 1
- 230000000840 anti-viral effect Effects 0.000 claims 1
- DNDCVAGJPBKION-DOPDSADYSA-N bombesin Chemical compound C([C@@H](C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCSC)C(N)=O)NC(=O)CNC(=O)[C@@H](NC(=O)[C@H](C)NC(=O)[C@H](CC=1NC2=CC=CC=C2C=1)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CC(N)=O)NC(=O)CNC(=O)[C@H](CC(C)C)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H]1NC(=O)CC1)C(C)C)C1=CN=CN1 DNDCVAGJPBKION-DOPDSADYSA-N 0.000 claims 1
- 230000024245 cell differentiation Effects 0.000 claims 1
- 108091008039 hormone receptors Proteins 0.000 claims 1
- 238000001794 hormone therapy Methods 0.000 claims 1
- 208000026278 immune system disease Diseases 0.000 claims 1
- 238000011534 incubation Methods 0.000 claims 1
- 229940100994 interleukin-7 Drugs 0.000 claims 1
- 230000010076 replication Effects 0.000 claims 1
- 230000003915 cell function Effects 0.000 abstract description 10
- 238000006243 chemical reaction Methods 0.000 abstract description 9
- 230000004060 metabolic process Effects 0.000 abstract description 5
- 230000001747 exhibiting effect Effects 0.000 abstract description 3
- 230000003116 impacting effect Effects 0.000 abstract description 3
- 230000019522 cellular metabolic process Effects 0.000 abstract description 2
- 239000000367 immunologic factor Substances 0.000 description 31
- 108090000623 proteins and genes Proteins 0.000 description 27
- 230000006870 function Effects 0.000 description 26
- 230000004044 response Effects 0.000 description 26
- 235000018102 proteins Nutrition 0.000 description 25
- 102000004169 proteins and genes Human genes 0.000 description 25
- 230000004069 differentiation Effects 0.000 description 24
- 102000037865 fusion proteins Human genes 0.000 description 22
- 108020001507 fusion proteins Proteins 0.000 description 22
- 108700033844 Pseudomonas aeruginosa toxA Proteins 0.000 description 19
- 210000002966 serum Anatomy 0.000 description 17
- 230000003993 interaction Effects 0.000 description 15
- 210000001519 tissue Anatomy 0.000 description 14
- 238000013459 approach Methods 0.000 description 12
- 230000004048 modification Effects 0.000 description 12
- 238000012986 modification Methods 0.000 description 12
- 201000005702 Pertussis Diseases 0.000 description 11
- 150000001413 amino acids Chemical class 0.000 description 11
- 230000001413 cellular effect Effects 0.000 description 11
- 206010008631 Cholera Diseases 0.000 description 10
- 230000004913 activation Effects 0.000 description 10
- 235000001014 amino acid Nutrition 0.000 description 10
- 231100000419 toxicity Toxicity 0.000 description 10
- 230000001988 toxicity Effects 0.000 description 10
- 231100000699 Bacterial toxin Toxicity 0.000 description 9
- 239000000688 bacterial toxin Substances 0.000 description 9
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 8
- 230000004075 alteration Effects 0.000 description 8
- 230000015572 biosynthetic process Effects 0.000 description 8
- 230000021615 conjugation Effects 0.000 description 8
- 239000003814 drug Substances 0.000 description 8
- 210000001616 monocyte Anatomy 0.000 description 8
- 239000000126 substance Substances 0.000 description 8
- 238000003786 synthesis reaction Methods 0.000 description 8
- ZKHQWZAMYRWXGA-KQYNXXCUSA-N Adenosine triphosphate Chemical compound C1=NC=2C(N)=NC=NC=2N1[C@@H]1O[C@H](COP(O)(=O)OP(O)(=O)OP(O)(O)=O)[C@@H](O)[C@H]1O ZKHQWZAMYRWXGA-KQYNXXCUSA-N 0.000 description 7
- ZKHQWZAMYRWXGA-UHFFFAOYSA-N Adenosine triphosphate Natural products C1=NC=2C(N)=NC=NC=2N1C1OC(COP(O)(=O)OP(O)(=O)OP(O)(O)=O)C(O)C1O ZKHQWZAMYRWXGA-UHFFFAOYSA-N 0.000 description 7
- 108090000288 Glycoproteins Proteins 0.000 description 7
- 102000003886 Glycoproteins Human genes 0.000 description 7
- 102000001253 Protein Kinase Human genes 0.000 description 7
- 230000001419 dependent effect Effects 0.000 description 7
- 230000002163 immunogen Effects 0.000 description 7
- 239000003112 inhibitor Substances 0.000 description 7
- 108060006633 protein kinase Proteins 0.000 description 7
- 230000002829 reductive effect Effects 0.000 description 7
- FALRKNHUBBKYCC-UHFFFAOYSA-N 2-(chloromethyl)pyridine-3-carbonitrile Chemical compound ClCC1=NC=CC=C1C#N FALRKNHUBBKYCC-UHFFFAOYSA-N 0.000 description 6
- 108010017213 Granulocyte-Macrophage Colony-Stimulating Factor Proteins 0.000 description 6
- NKANXQFJJICGDU-QPLCGJKRSA-N Tamoxifen Chemical compound C=1C=CC=CC=1C(/CC)=C(C=1C=CC(OCCN(C)C)=CC=1)/C1=CC=CC=C1 NKANXQFJJICGDU-QPLCGJKRSA-N 0.000 description 6
- 210000000805 cytoplasm Anatomy 0.000 description 6
- 210000000987 immune system Anatomy 0.000 description 6
- 230000002637 immunotoxin Effects 0.000 description 6
- 231100000608 immunotoxin Toxicity 0.000 description 6
- 229940051026 immunotoxin Drugs 0.000 description 6
- 230000006698 induction Effects 0.000 description 6
- 230000003834 intracellular effect Effects 0.000 description 6
- 210000004698 lymphocyte Anatomy 0.000 description 6
- 230000009467 reduction Effects 0.000 description 6
- 229940014800 succinic anhydride Drugs 0.000 description 6
- JWDFQMWEFLOOED-UHFFFAOYSA-N (2,5-dioxopyrrolidin-1-yl) 3-(pyridin-2-yldisulfanyl)propanoate Chemical compound O=C1CCC(=O)N1OC(=O)CCSSC1=CC=CC=N1 JWDFQMWEFLOOED-UHFFFAOYSA-N 0.000 description 5
- 108010053187 Diphtheria Toxin Proteins 0.000 description 5
- 108010017080 Granulocyte Colony-Stimulating Factor Proteins 0.000 description 5
- 102000004269 Granulocyte Colony-Stimulating Factor Human genes 0.000 description 5
- XKMLYUALXHKNFT-UUOKFMHZSA-N Guanosine-5'-triphosphate Chemical compound C1=2NC(N)=NC(=O)C=2N=CN1[C@@H]1O[C@H](COP(O)(=O)OP(O)(=O)OP(O)(O)=O)[C@@H](O)[C@H]1O XKMLYUALXHKNFT-UUOKFMHZSA-N 0.000 description 5
- KDXKERNSBIXSRK-YFKPBYRVSA-N L-lysine Chemical compound NCCCC[C@H](N)C(O)=O KDXKERNSBIXSRK-YFKPBYRVSA-N 0.000 description 5
- 102000043276 Oncogene Human genes 0.000 description 5
- 108700020796 Oncogene Proteins 0.000 description 5
- 230000030741 antigen processing and presentation Effects 0.000 description 5
- 230000008901 benefit Effects 0.000 description 5
- 230000030833 cell death Effects 0.000 description 5
- 230000033077 cellular process Effects 0.000 description 5
- 238000002512 chemotherapy Methods 0.000 description 5
- 125000004122 cyclic group Chemical group 0.000 description 5
- 235000018977 lysine Nutrition 0.000 description 5
- 125000003588 lysine group Chemical group [H]N([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])(N([H])[H])C(*)=O 0.000 description 5
- 238000004519 manufacturing process Methods 0.000 description 5
- 238000007799 mixed lymphocyte reaction assay Methods 0.000 description 5
- 238000011275 oncology therapy Methods 0.000 description 5
- 230000001629 suppression Effects 0.000 description 5
- 238000012360 testing method Methods 0.000 description 5
- 229940124597 therapeutic agent Drugs 0.000 description 5
- 231100000331 toxic Toxicity 0.000 description 5
- 230000002588 toxic effect Effects 0.000 description 5
- 230000001960 triggered effect Effects 0.000 description 5
- 208000026310 Breast neoplasm Diseases 0.000 description 4
- 206010012735 Diarrhoea Diseases 0.000 description 4
- 102000016607 Diphtheria Toxin Human genes 0.000 description 4
- 102000034353 G alpha subunit Human genes 0.000 description 4
- 108091006099 G alpha subunit Proteins 0.000 description 4
- NTYJJOPFIAHURM-UHFFFAOYSA-N Histamine Chemical compound NCCC1=CN=CN1 NTYJJOPFIAHURM-UHFFFAOYSA-N 0.000 description 4
- 241000282412 Homo Species 0.000 description 4
- KDXKERNSBIXSRK-UHFFFAOYSA-N Lysine Natural products NCCCCC(N)C(O)=O KDXKERNSBIXSRK-UHFFFAOYSA-N 0.000 description 4
- 239000004472 Lysine Substances 0.000 description 4
- 241000699670 Mus sp. Species 0.000 description 4
- 102000035195 Peptidases Human genes 0.000 description 4
- 108010039491 Ricin Proteins 0.000 description 4
- 230000009471 action Effects 0.000 description 4
- 125000000539 amino acid group Chemical group 0.000 description 4
- 230000001147 anti-toxic effect Effects 0.000 description 4
- 201000011510 cancer Diseases 0.000 description 4
- 230000003197 catalytic effect Effects 0.000 description 4
- 238000003776 cleavage reaction Methods 0.000 description 4
- 238000012217 deletion Methods 0.000 description 4
- 230000037430 deletion Effects 0.000 description 4
- VHJLVAABSRFDPM-QWWZWVQMSA-N dithiothreitol Chemical compound SC[C@@H](O)[C@H](O)CS VHJLVAABSRFDPM-QWWZWVQMSA-N 0.000 description 4
- 231100000371 dose-limiting toxicity Toxicity 0.000 description 4
- 210000003527 eukaryotic cell Anatomy 0.000 description 4
- QPJBWNIQKHGLAU-IQZHVAEDSA-N ganglioside GM1 Chemical compound O[C@@H]1[C@@H](O)[C@H](OC[C@H](NC(=O)CCCCCCCCCCCCCCCCC)[C@H](O)\C=C\CCCCCCCCCCCCC)O[C@H](CO)[C@H]1O[C@H]1[C@H](O)[C@@H](O[C@]2(O[C@H]([C@H](NC(C)=O)[C@@H](O)C2)[C@H](O)[C@H](O)CO)C(O)=O)[C@@H](O[C@H]2[C@@H]([C@@H](O[C@H]3[C@@H]([C@@H](O)[C@@H](O)[C@@H](CO)O3)O)[C@@H](O)[C@@H](CO)O2)NC(C)=O)[C@@H](CO)O1 QPJBWNIQKHGLAU-IQZHVAEDSA-N 0.000 description 4
- 230000012010 growth Effects 0.000 description 4
- 230000028993 immune response Effects 0.000 description 4
- NOESYZHRGYRDHS-UHFFFAOYSA-N insulin Chemical compound N1C(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(NC(=O)CN)C(C)CC)CSSCC(C(NC(CO)C(=O)NC(CC(C)C)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CCC(N)=O)C(=O)NC(CC(C)C)C(=O)NC(CCC(O)=O)C(=O)NC(CC(N)=O)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CSSCC(NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2C=CC(O)=CC=2)NC(=O)C(CC(C)C)NC(=O)C(C)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2NC=NC=2)NC(=O)C(CO)NC(=O)CNC2=O)C(=O)NCC(=O)NC(CCC(O)=O)C(=O)NC(CCCNC(N)=N)C(=O)NCC(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC(O)=CC=3)C(=O)NC(C(C)O)C(=O)N3C(CCC3)C(=O)NC(CCCCN)C(=O)NC(C)C(O)=O)C(=O)NC(CC(N)=O)C(O)=O)=O)NC(=O)C(C(C)CC)NC(=O)C(CO)NC(=O)C(C(C)O)NC(=O)C1CSSCC2NC(=O)C(CC(C)C)NC(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CC(N)=O)NC(=O)C(NC(=O)C(N)CC=1C=CC=CC=1)C(C)C)CC1=CN=CN1 NOESYZHRGYRDHS-UHFFFAOYSA-N 0.000 description 4
- 210000000265 leukocyte Anatomy 0.000 description 4
- 230000000670 limiting effect Effects 0.000 description 4
- 210000002540 macrophage Anatomy 0.000 description 4
- 230000001613 neoplastic effect Effects 0.000 description 4
- 230000002062 proliferating effect Effects 0.000 description 4
- 230000002035 prolonged effect Effects 0.000 description 4
- 231100000654 protein toxin Toxicity 0.000 description 4
- 108700042226 ras Genes Proteins 0.000 description 4
- 230000007017 scission Effects 0.000 description 4
- 230000028327 secretion Effects 0.000 description 4
- 230000019491 signal transduction Effects 0.000 description 4
- 239000011780 sodium chloride Substances 0.000 description 4
- QXZBMSIDSOZZHK-DOPDSADYSA-N 31362-50-2 Chemical compound C([C@@H](C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCSC)C(N)=O)NC(=O)CNC(=O)[C@@H](NC(=O)[C@H](C)NC(=O)[C@H](CC=1C2=CC=CC=C2NC=1)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CC(N)=O)NC(=O)CNC(=O)[C@H](CC(C)C)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H]1NC(=O)CC1)C(C)C)C1=CNC=N1 QXZBMSIDSOZZHK-DOPDSADYSA-N 0.000 description 3
- 206010006187 Breast cancer Diseases 0.000 description 3
- 102000007644 Colony-Stimulating Factors Human genes 0.000 description 3
- 102100031334 Elongation factor 2 Human genes 0.000 description 3
- 241000196324 Embryophyta Species 0.000 description 3
- 108700004714 Gelonium multiflorum GEL Proteins 0.000 description 3
- 102000006354 HLA-DR Antigens Human genes 0.000 description 3
- 108010058597 HLA-DR Antigens Proteins 0.000 description 3
- 102000000589 Interleukin-1 Human genes 0.000 description 3
- 108010002352 Interleukin-1 Proteins 0.000 description 3
- 108010002386 Interleukin-3 Proteins 0.000 description 3
- 102000000646 Interleukin-3 Human genes 0.000 description 3
- 108010046938 Macrophage Colony-Stimulating Factor Proteins 0.000 description 3
- 102000007651 Macrophage Colony-Stimulating Factor Human genes 0.000 description 3
- 206010027476 Metastases Diseases 0.000 description 3
- 108010077519 Peptide Elongation Factor 2 Proteins 0.000 description 3
- 238000009825 accumulation Methods 0.000 description 3
- 230000033228 biological regulation Effects 0.000 description 3
- 210000004369 blood Anatomy 0.000 description 3
- 239000008280 blood Substances 0.000 description 3
- 230000022534 cell killing Effects 0.000 description 3
- 238000007385 chemical modification Methods 0.000 description 3
- 238000010276 construction Methods 0.000 description 3
- 238000013461 design Methods 0.000 description 3
- 206010013023 diphtheria Diseases 0.000 description 3
- 238000009826 distribution Methods 0.000 description 3
- 229940079593 drug Drugs 0.000 description 3
- 210000000981 epithelium Anatomy 0.000 description 3
- 230000001976 improved effect Effects 0.000 description 3
- 230000002779 inactivation Effects 0.000 description 3
- 238000002347 injection Methods 0.000 description 3
- 239000007924 injection Substances 0.000 description 3
- 230000010534 mechanism of action Effects 0.000 description 3
- 231100000252 nontoxic Toxicity 0.000 description 3
- 230000003000 nontoxic effect Effects 0.000 description 3
- 238000006116 polymerization reaction Methods 0.000 description 3
- 238000010188 recombinant method Methods 0.000 description 3
- 230000004936 stimulating effect Effects 0.000 description 3
- 230000000638 stimulation Effects 0.000 description 3
- 230000035322 succinylation Effects 0.000 description 3
- 238000010613 succinylation reaction Methods 0.000 description 3
- 229960001603 tamoxifen Drugs 0.000 description 3
- 230000005945 translocation Effects 0.000 description 3
- 230000003827 upregulation 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
- LLXVXPPXELIDGQ-UHFFFAOYSA-N (2,5-dioxopyrrolidin-1-yl) 3-(2,5-dioxopyrrol-1-yl)benzoate Chemical compound C=1C=CC(N2C(C=CC2=O)=O)=CC=1C(=O)ON1C(=O)CCC1=O LLXVXPPXELIDGQ-UHFFFAOYSA-N 0.000 description 2
- BQWBEDSJTMWJAE-UHFFFAOYSA-N (2,5-dioxopyrrolidin-1-yl) 4-[(2-iodoacetyl)amino]benzoate Chemical compound C1=CC(NC(=O)CI)=CC=C1C(=O)ON1C(=O)CCC1=O BQWBEDSJTMWJAE-UHFFFAOYSA-N 0.000 description 2
- PMJWDPGOWBRILU-UHFFFAOYSA-N (2,5-dioxopyrrolidin-1-yl) 4-[4-(2,5-dioxopyrrol-1-yl)phenyl]butanoate Chemical compound O=C1CCC(=O)N1OC(=O)CCCC(C=C1)=CC=C1N1C(=O)C=CC1=O PMJWDPGOWBRILU-UHFFFAOYSA-N 0.000 description 2
- DIYPCWKHSODVAP-UHFFFAOYSA-N 1-[3-(2,5-dioxopyrrol-1-yl)benzoyl]oxy-2,5-dioxopyrrolidine-3-sulfonic acid Chemical compound O=C1C(S(=O)(=O)O)CC(=O)N1OC(=O)C1=CC=CC(N2C(C=CC2=O)=O)=C1 DIYPCWKHSODVAP-UHFFFAOYSA-N 0.000 description 2
- FPKVOQKZMBDBKP-UHFFFAOYSA-N 1-[4-[(2,5-dioxopyrrol-1-yl)methyl]cyclohexanecarbonyl]oxy-2,5-dioxopyrrolidine-3-sulfonic acid Chemical compound O=C1C(S(=O)(=O)O)CC(=O)N1OC(=O)C1CCC(CN2C(C=CC2=O)=O)CC1 FPKVOQKZMBDBKP-UHFFFAOYSA-N 0.000 description 2
- VHYRLCJMMJQUBY-UHFFFAOYSA-N 1-[4-[4-(2,5-dioxopyrrol-1-yl)phenyl]butanoyloxy]-2,5-dioxopyrrolidine-3-sulfonic acid Chemical compound O=C1C(S(=O)(=O)O)CC(=O)N1OC(=O)CCCC1=CC=C(N2C(C=CC2=O)=O)C=C1 VHYRLCJMMJQUBY-UHFFFAOYSA-N 0.000 description 2
- VOXZDWNPVJITMN-ZBRFXRBCSA-N 17β-estradiol Chemical compound OC1=CC=C2[C@H]3CC[C@](C)([C@H](CC4)O)[C@@H]4[C@@H]3CCC2=C1 VOXZDWNPVJITMN-ZBRFXRBCSA-N 0.000 description 2
- AYKYXWQEBUNJCN-UHFFFAOYSA-N 3-methylfuran-2,5-dione Chemical compound CC1=CC(=O)OC1=O AYKYXWQEBUNJCN-UHFFFAOYSA-N 0.000 description 2
- 208000030507 AIDS Diseases 0.000 description 2
- 108010066676 Abrin Proteins 0.000 description 2
- 108010088751 Albumins Proteins 0.000 description 2
- 102000009027 Albumins Human genes 0.000 description 2
- 108020000948 Antisense Oligonucleotides Proteins 0.000 description 2
- 208000023275 Autoimmune disease Diseases 0.000 description 2
- 238000011725 BALB/c mouse Methods 0.000 description 2
- 241000894006 Bacteria Species 0.000 description 2
- 102000000584 Calmodulin Human genes 0.000 description 2
- 108010041952 Calmodulin Proteins 0.000 description 2
- 102000005600 Cathepsins Human genes 0.000 description 2
- 108010084457 Cathepsins Proteins 0.000 description 2
- PMATZTZNYRCHOR-CGLBZJNRSA-N Cyclosporin A Chemical compound CC[C@@H]1NC(=O)[C@H]([C@H](O)[C@H](C)C\C=C\C)N(C)C(=O)[C@H](C(C)C)N(C)C(=O)[C@H](CC(C)C)N(C)C(=O)[C@H](CC(C)C)N(C)C(=O)[C@@H](C)NC(=O)[C@H](C)NC(=O)[C@H](CC(C)C)N(C)C(=O)[C@H](C(C)C)NC(=O)[C@H](CC(C)C)N(C)C(=O)CN(C)C1=O PMATZTZNYRCHOR-CGLBZJNRSA-N 0.000 description 2
- 229930105110 Cyclosporin A Natural products 0.000 description 2
- 108010036949 Cyclosporine Proteins 0.000 description 2
- 206010059866 Drug resistance Diseases 0.000 description 2
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 2
- OHCQJHSOBUTRHG-KGGHGJDLSA-N FORSKOLIN Chemical compound O=C([C@@]12O)C[C@](C)(C=C)O[C@]1(C)[C@@H](OC(=O)C)[C@@H](O)[C@@H]1[C@]2(C)[C@@H](O)CCC1(C)C OHCQJHSOBUTRHG-KGGHGJDLSA-N 0.000 description 2
- WHUUTDBJXJRKMK-UHFFFAOYSA-N Glutamic acid Natural products OC(=O)C(N)CCC(O)=O WHUUTDBJXJRKMK-UHFFFAOYSA-N 0.000 description 2
- 102000004877 Insulin Human genes 0.000 description 2
- 108090001061 Insulin Proteins 0.000 description 2
- 102000010789 Interleukin-2 Receptors Human genes 0.000 description 2
- 108010038453 Interleukin-2 Receptors Proteins 0.000 description 2
- 102000000704 Interleukin-7 Human genes 0.000 description 2
- 102000015696 Interleukins Human genes 0.000 description 2
- 108010063738 Interleukins Proteins 0.000 description 2
- 108090000862 Ion Channels Proteins 0.000 description 2
- 102000004310 Ion Channels Human genes 0.000 description 2
- HNDVDQJCIGZPNO-YFKPBYRVSA-N L-histidine Chemical compound OC(=O)[C@@H](N)CC1=CN=CN1 HNDVDQJCIGZPNO-YFKPBYRVSA-N 0.000 description 2
- QIVBCDIJIAJPQS-VIFPVBQESA-N L-tryptophane Chemical compound C1=CC=C2C(C[C@H](N)C(O)=O)=CNC2=C1 QIVBCDIJIAJPQS-VIFPVBQESA-N 0.000 description 2
- 102000043131 MHC class II family Human genes 0.000 description 2
- 108091054438 MHC class II family Proteins 0.000 description 2
- 231100000742 Plant toxin Toxicity 0.000 description 2
- 241000607768 Shigella Species 0.000 description 2
- 206010052779 Transplant rejections Diseases 0.000 description 2
- QIVBCDIJIAJPQS-UHFFFAOYSA-N Tryptophan Natural products C1=CC=C2C(CC(N)C(O)=O)=CNC2=C1 QIVBCDIJIAJPQS-UHFFFAOYSA-N 0.000 description 2
- 230000002159 abnormal effect Effects 0.000 description 2
- 201000009840 acute diarrhea Diseases 0.000 description 2
- OIRDTQYFTABQOQ-KQYNXXCUSA-N adenosine Chemical compound C1=NC=2C(N)=NC=NC=2N1[C@@H]1O[C@H](CO)[C@@H](O)[C@H]1O OIRDTQYFTABQOQ-KQYNXXCUSA-N 0.000 description 2
- 230000002411 adverse Effects 0.000 description 2
- 239000000556 agonist Substances 0.000 description 2
- 230000003321 amplification Effects 0.000 description 2
- 125000000129 anionic group Chemical group 0.000 description 2
- 239000002246 antineoplastic agent Substances 0.000 description 2
- 239000000074 antisense oligonucleotide Substances 0.000 description 2
- 238000012230 antisense oligonucleotides Methods 0.000 description 2
- 230000004071 biological effect Effects 0.000 description 2
- 230000015556 catabolic process Effects 0.000 description 2
- 210000000170 cell membrane Anatomy 0.000 description 2
- 229960001265 ciclosporin Drugs 0.000 description 2
- 230000000112 colonic effect Effects 0.000 description 2
- 238000002648 combination therapy Methods 0.000 description 2
- 150000001875 compounds Chemical class 0.000 description 2
- 239000000470 constituent Substances 0.000 description 2
- 230000008878 coupling Effects 0.000 description 2
- 238000010168 coupling process Methods 0.000 description 2
- 238000005859 coupling reaction Methods 0.000 description 2
- 229930182912 cyclosporin Natural products 0.000 description 2
- 230000034994 death Effects 0.000 description 2
- 238000006731 degradation reaction Methods 0.000 description 2
- 201000010099 disease Diseases 0.000 description 2
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 2
- 230000009977 dual effect Effects 0.000 description 2
- 230000007515 enzymatic degradation Effects 0.000 description 2
- 230000009144 enzymatic modification Effects 0.000 description 2
- 102000015694 estrogen receptors Human genes 0.000 description 2
- 108010038795 estrogen receptors Proteins 0.000 description 2
- 230000029142 excretion Effects 0.000 description 2
- 239000002095 exotoxin Substances 0.000 description 2
- 231100000776 exotoxin Toxicity 0.000 description 2
- 238000002474 experimental method Methods 0.000 description 2
- 230000005714 functional activity Effects 0.000 description 2
- 150000002270 gangliosides Chemical class 0.000 description 2
- 238000010353 genetic engineering Methods 0.000 description 2
- 235000013922 glutamic acid Nutrition 0.000 description 2
- 239000004220 glutamic acid Substances 0.000 description 2
- RWSXRVCMGQZWBV-WDSKDSINSA-N glutathione Chemical compound OC(=O)[C@@H](N)CCC(=O)N[C@@H](CS)C(=O)NCC(O)=O RWSXRVCMGQZWBV-WDSKDSINSA-N 0.000 description 2
- 230000002440 hepatic effect Effects 0.000 description 2
- 210000003494 hepatocyte Anatomy 0.000 description 2
- 229960001340 histamine Drugs 0.000 description 2
- HNDVDQJCIGZPNO-UHFFFAOYSA-N histidine Natural products OC(=O)C(N)CC1=CN=CN1 HNDVDQJCIGZPNO-UHFFFAOYSA-N 0.000 description 2
- 230000006303 immediate early viral mRNA transcription Effects 0.000 description 2
- 230000001900 immune effect Effects 0.000 description 2
- 229940125396 insulin Drugs 0.000 description 2
- 238000001990 intravenous administration Methods 0.000 description 2
- 210000003734 kidney Anatomy 0.000 description 2
- 230000002147 killing effect Effects 0.000 description 2
- 210000004185 liver Anatomy 0.000 description 2
- 230000002132 lysosomal effect Effects 0.000 description 2
- 230000036210 malignancy Effects 0.000 description 2
- 230000035800 maturation Effects 0.000 description 2
- 210000003622 mature neutrocyte Anatomy 0.000 description 2
- 230000009401 metastasis Effects 0.000 description 2
- 239000000203 mixture Substances 0.000 description 2
- 238000010172 mouse model Methods 0.000 description 2
- 238000003199 nucleic acid amplification method Methods 0.000 description 2
- 239000003123 plant toxin Substances 0.000 description 2
- 108700028325 pokeweed antiviral Proteins 0.000 description 2
- OXCMYAYHXIHQOA-UHFFFAOYSA-N potassium;[2-butyl-5-chloro-3-[[4-[2-(1,2,4-triaza-3-azanidacyclopenta-1,4-dien-5-yl)phenyl]phenyl]methyl]imidazol-4-yl]methanol Chemical compound [K+].CCCCC1=NC(Cl)=C(CO)N1CC1=CC=C(C=2C(=CC=CC=2)C2=N[N-]N=N2)C=C1 OXCMYAYHXIHQOA-UHFFFAOYSA-N 0.000 description 2
- 230000009145 protein modification Effects 0.000 description 2
- 230000005855 radiation Effects 0.000 description 2
- -1 radioisotopes Substances 0.000 description 2
- 229940121896 radiopharmaceutical Drugs 0.000 description 2
- 239000012217 radiopharmaceutical Substances 0.000 description 2
- 230000002799 radiopharmaceutical effect Effects 0.000 description 2
- 230000009257 reactivity Effects 0.000 description 2
- 238000011084 recovery Methods 0.000 description 2
- 238000011160 research Methods 0.000 description 2
- 230000019254 respiratory burst Effects 0.000 description 2
- 238000002741 site-directed mutagenesis Methods 0.000 description 2
- HHSGWIABCIVPJT-UHFFFAOYSA-M sodium;1-[4-[(2-iodoacetyl)amino]benzoyl]oxy-2,5-dioxopyrrolidine-3-sulfonate Chemical compound [Na+].O=C1C(S(=O)(=O)[O-])CC(=O)N1OC(=O)C1=CC=C(NC(=O)CI)C=C1 HHSGWIABCIVPJT-UHFFFAOYSA-M 0.000 description 2
- 239000007858 starting material Substances 0.000 description 2
- JJAHTWIKCUJRDK-UHFFFAOYSA-N succinimidyl 4-(N-maleimidomethyl)cyclohexane-1-carboxylate Chemical compound C1CC(CN2C(C=CC2=O)=O)CCC1C(=O)ON1C(=O)CCC1=O JJAHTWIKCUJRDK-UHFFFAOYSA-N 0.000 description 2
- 230000004083 survival effect Effects 0.000 description 2
- 231100001274 therapeutic index Toxicity 0.000 description 2
- 125000003396 thiol group Chemical group [H]S* 0.000 description 2
- 230000004565 tumor cell growth Effects 0.000 description 2
- YFGBQHOOROIVKG-BHDDXSALSA-N (2R)-2-[[(2R)-2-[[2-[[2-[[(2S)-2-amino-3-(4-hydroxyphenyl)propanoyl]amino]acetyl]amino]acetyl]amino]-3-phenylpropanoyl]amino]-4-methylsulfanylbutanoic acid Chemical compound C([C@H](C(=O)N[C@H](CCSC)C(O)=O)NC(=O)CNC(=O)CNC(=O)[C@@H](N)CC=1C=CC(O)=CC=1)C1=CC=CC=C1 YFGBQHOOROIVKG-BHDDXSALSA-N 0.000 description 1
- MTCFGRXMJLQNBG-REOHCLBHSA-N (2S)-2-Amino-3-hydroxypropansäure Chemical compound OC[C@H](N)C(O)=O MTCFGRXMJLQNBG-REOHCLBHSA-N 0.000 description 1
- RWBLWXCGQLZKLK-USVTTYPOSA-N (2s)-2-[(2-aminoacetyl)amino]-n-[(2s)-1-[[(2s)-1-[[(2s)-1-[[(2s)-1-[[2-[[(2s)-1-[[(2s)-1-[[(2s)-1-amino-4-methylsulfanyl-1-oxobutan-2-yl]amino]-4-methyl-1-oxopentan-2-yl]amino]-3-(1h-imidazol-5-yl)-1-oxopropan-2-yl]amino]-2-oxoethyl]amino]-3-methyl-1-oxob Chemical compound C([C@@H](C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCSC)C(N)=O)NC(=O)CNC(=O)[C@@H](NC(=O)[C@H](C)NC(=O)[C@H](CC=1C2=CC=CC=C2NC=1)NC(=O)[C@H](CC=1NC=NC=1)NC(=O)[C@H](CC(N)=O)NC(=O)CN)C(C)C)C1=CN=CN1 RWBLWXCGQLZKLK-USVTTYPOSA-N 0.000 description 1
- YPFNACALNKVZNK-MFNIMNRCSA-N (2s)-2-[(2-aminoacetyl)amino]-n-[(2s)-1-[[(2s)-1-[[(2s)-1-[[(2s,3r)-1-[[2-[[(2s)-1-[[(2s)-1-[[(2s)-1-amino-4-methylsulfanyl-1-oxobutan-2-yl]amino]-1-oxo-3-phenylpropan-2-yl]amino]-3-(1h-imidazol-5-yl)-1-oxopropan-2-yl]amino]-2-oxoethyl]amino]-3-hydroxy-1- Chemical compound C([C@@H](C(=O)N[C@@H](CCSC)C(N)=O)NC(=O)[C@H](CC=1NC=NC=1)NC(=O)CNC(=O)[C@@H](NC(=O)[C@H](C)NC(=O)[C@H](CC=1C2=CC=CC=C2NC=1)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC(N)=O)NC(=O)CN)[C@@H](C)O)C1=CC=CC=C1 YPFNACALNKVZNK-MFNIMNRCSA-N 0.000 description 1
- CLLFEJLEDNXZNR-UUOKFMHZSA-N (4ar,6r,7r,7as)-6-(6-amino-8-chloropurin-9-yl)-2-hydroxy-2-oxo-4a,6,7,7a-tetrahydro-4h-furo[3,2-d][1,3,2]dioxaphosphinin-7-ol Chemical compound C([C@H]1O2)OP(O)(=O)O[C@H]1[C@@H](O)[C@@H]2N1C(N=CN=C2N)=C2N=C1Cl CLLFEJLEDNXZNR-UUOKFMHZSA-N 0.000 description 1
- ZIIUUSVHCHPIQD-UHFFFAOYSA-N 2,4,6-trimethyl-N-[3-(trifluoromethyl)phenyl]benzenesulfonamide Chemical compound CC1=CC(C)=CC(C)=C1S(=O)(=O)NC1=CC=CC(C(F)(F)F)=C1 ZIIUUSVHCHPIQD-UHFFFAOYSA-N 0.000 description 1
- FPWYUEKIRUBHML-LGVAUZIVSA-N 2-amino-9-[(2r,3r,4s,5r)-3,4-dihydroxy-5-(hydroxymethyl)oxolan-2-yl]-3h-purin-6-one;phosphoric acid Chemical compound OP(O)(O)=O.OP(O)(O)=O.C1=2NC(N)=NC(=O)C=2N=CN1[C@@H]1O[C@H](CO)[C@@H](O)[C@H]1O FPWYUEKIRUBHML-LGVAUZIVSA-N 0.000 description 1
- DJBRKGZFUXKLKO-UHFFFAOYSA-N 3-(pyridin-2-yldisulfanyl)propanoic acid Chemical compound OC(=O)CCSSC1=CC=CC=N1 DJBRKGZFUXKLKO-UHFFFAOYSA-N 0.000 description 1
- HVCOBJNICQPDBP-UHFFFAOYSA-N 3-[3-[3,5-dihydroxy-6-methyl-4-(3,4,5-trihydroxy-6-methyloxan-2-yl)oxyoxan-2-yl]oxydecanoyloxy]decanoic acid;hydrate Chemical compound O.OC1C(OC(CC(=O)OC(CCCCCCC)CC(O)=O)CCCCCCC)OC(C)C(O)C1OC1C(O)C(O)C(O)C(C)O1 HVCOBJNICQPDBP-UHFFFAOYSA-N 0.000 description 1
- 108091006112 ATPases Proteins 0.000 description 1
- 102000057290 Adenosine Triphosphatases Human genes 0.000 description 1
- 102100033312 Alpha-2-macroglobulin Human genes 0.000 description 1
- 102000006306 Antigen Receptors Human genes 0.000 description 1
- 108010083359 Antigen Receptors Proteins 0.000 description 1
- 101000605172 Aspergillus niger (strain CBS 513.88 / FGSC A1513) Probable endopolygalacturonase E Proteins 0.000 description 1
- 101000605171 Aspergillus niger Endopolygalacturonase E Proteins 0.000 description 1
- 108090001008 Avidin Proteins 0.000 description 1
- 241000193738 Bacillus anthracis Species 0.000 description 1
- 108010006654 Bleomycin Proteins 0.000 description 1
- 102000004506 Blood Proteins Human genes 0.000 description 1
- 108010017384 Blood Proteins Proteins 0.000 description 1
- 101000941281 Bos taurus Gastric triacylglycerol lipase Proteins 0.000 description 1
- 239000002126 C01EB10 - Adenosine Substances 0.000 description 1
- 201000009030 Carcinoma Diseases 0.000 description 1
- 102000014914 Carrier Proteins Human genes 0.000 description 1
- 108010001857 Cell Surface Receptors Proteins 0.000 description 1
- 108091006146 Channels Proteins 0.000 description 1
- 108091035707 Consensus sequence Proteins 0.000 description 1
- 239000000055 Corticotropin-Releasing Hormone Substances 0.000 description 1
- 239000004971 Cross linker Substances 0.000 description 1
- IVOMOUWHDPKRLL-KQYNXXCUSA-N Cyclic adenosine monophosphate Chemical compound C([C@H]1O2)OP(O)(=O)O[C@H]1[C@@H](O)[C@@H]2N1C(N=CN=C2N)=C2N=C1 IVOMOUWHDPKRLL-KQYNXXCUSA-N 0.000 description 1
- 108700020911 DNA-Binding Proteins Proteins 0.000 description 1
- 102000052510 DNA-Binding Proteins Human genes 0.000 description 1
- SUZLHDUTVMZSEV-UHFFFAOYSA-N Deoxycoleonol Natural products C12C(=O)CC(C)(C=C)OC2(C)C(OC(=O)C)C(O)C2C1(C)C(O)CCC2(C)C SUZLHDUTVMZSEV-UHFFFAOYSA-N 0.000 description 1
- 238000002965 ELISA Methods 0.000 description 1
- 108010092408 Eosinophil Peroxidase Proteins 0.000 description 1
- 102000003951 Erythropoietin Human genes 0.000 description 1
- 102100031939 Erythropoietin Human genes 0.000 description 1
- 108090000394 Erythropoietin Proteins 0.000 description 1
- 101150021185 FGF gene Proteins 0.000 description 1
- 239000004606 Fillers/Extenders Substances 0.000 description 1
- 102000012673 Follicle Stimulating Hormone Human genes 0.000 description 1
- 108010079345 Follicle Stimulating Hormone Proteins 0.000 description 1
- 102000034354 Gi proteins Human genes 0.000 description 1
- 108091006101 Gi proteins Proteins 0.000 description 1
- WQZGKKKJIJFFOK-GASJEMHNSA-N Glucose Natural products OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 1
- 108010024636 Glutathione Proteins 0.000 description 1
- 229930186217 Glycolipid Natural products 0.000 description 1
- 102000004457 Granulocyte-Macrophage Colony-Stimulating Factor Human genes 0.000 description 1
- QIFOTGBMWFMXRC-UHFFFAOYSA-N Holotoxin A Natural products OC1C(OC)C(O)C(CO)OC1OC1C(O)C(OC2C(OCC(C2O)OC2C(C(O)C(OC3C(C(O)C(O)CO3)O)C(C)O2)O)OC2C(C3C(C=4C(C5(CC(=O)C6C5(C(OC6(CCCC(C)(C)O)C)=O)CC=4)C)CC3)(C)CC2)(C)C)OC(CO)C1O QIFOTGBMWFMXRC-UHFFFAOYSA-N 0.000 description 1
- 101001012157 Homo sapiens Receptor tyrosine-protein kinase erbB-2 Proteins 0.000 description 1
- 101000818522 Homo sapiens fMet-Leu-Phe receptor Proteins 0.000 description 1
- 240000005979 Hordeum vulgare Species 0.000 description 1
- 235000007340 Hordeum vulgare Nutrition 0.000 description 1
- 239000000854 Human Growth Hormone Substances 0.000 description 1
- 102000002265 Human Growth Hormone Human genes 0.000 description 1
- 108010000521 Human Growth Hormone Proteins 0.000 description 1
- 108010021625 Immunoglobulin Fragments Proteins 0.000 description 1
- 102000008394 Immunoglobulin Fragments Human genes 0.000 description 1
- 206010062016 Immunosuppression Diseases 0.000 description 1
- 206010061218 Inflammation Diseases 0.000 description 1
- 102100026688 Interferon epsilon Human genes 0.000 description 1
- 101710147309 Interferon epsilon Proteins 0.000 description 1
- 102000008070 Interferon-gamma Human genes 0.000 description 1
- 108010074328 Interferon-gamma Proteins 0.000 description 1
- 102000014150 Interferons Human genes 0.000 description 1
- 108010050904 Interferons Proteins 0.000 description 1
- 108090000978 Interleukin-4 Proteins 0.000 description 1
- 102000004388 Interleukin-4 Human genes 0.000 description 1
- SHGAZHPCJJPHSC-NUEINMDLSA-N Isotretinoin Chemical compound OC(=O)C=C(C)/C=C/C=C(C)C=CC1=C(C)CCCC1(C)C SHGAZHPCJJPHSC-NUEINMDLSA-N 0.000 description 1
- XUJNEKJLAYXESH-REOHCLBHSA-N L-Cysteine Chemical compound SC[C@H](N)C(O)=O XUJNEKJLAYXESH-REOHCLBHSA-N 0.000 description 1
- AYFVYJQAPQTCCC-GBXIJSLDSA-N L-threonine Chemical compound C[C@@H](O)[C@H](N)C(O)=O AYFVYJQAPQTCCC-GBXIJSLDSA-N 0.000 description 1
- OUYCCCASQSFEME-QMMMGPOBSA-N L-tyrosine Chemical compound OC(=O)[C@@H](N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-QMMMGPOBSA-N 0.000 description 1
- 102000007330 LDL Lipoproteins Human genes 0.000 description 1
- 108010007622 LDL Lipoproteins Proteins 0.000 description 1
- 108090001090 Lectins Proteins 0.000 description 1
- 102000004856 Lectins Human genes 0.000 description 1
- 102000009151 Luteinizing Hormone Human genes 0.000 description 1
- 108010073521 Luteinizing Hormone Proteins 0.000 description 1
- 206010025280 Lymphocytosis Diseases 0.000 description 1
- 102000008072 Lymphokines Human genes 0.000 description 1
- 108010074338 Lymphokines Proteins 0.000 description 1
- 241000124008 Mammalia Species 0.000 description 1
- 239000000637 Melanocyte-Stimulating Hormone Substances 0.000 description 1
- 108010007013 Melanocyte-Stimulating Hormones Proteins 0.000 description 1
- 102400000988 Met-enkephalin Human genes 0.000 description 1
- 108010042237 Methionine Enkephalin Proteins 0.000 description 1
- 241001529936 Murinae Species 0.000 description 1
- 241000699666 Mus <mouse, genus> Species 0.000 description 1
- 102400001084 Neuromedin-B Human genes 0.000 description 1
- 101800001639 Neuromedin-B Proteins 0.000 description 1
- 101800001638 Neuromedin-C Proteins 0.000 description 1
- 102400001103 Neurotensin Human genes 0.000 description 1
- 101800001814 Neurotensin Proteins 0.000 description 1
- 229910019142 PO4 Inorganic materials 0.000 description 1
- 102000015731 Peptide Hormones Human genes 0.000 description 1
- 108010038988 Peptide Hormones Proteins 0.000 description 1
- 206010057249 Phagocytosis Diseases 0.000 description 1
- 102000015439 Phospholipases Human genes 0.000 description 1
- 108010064785 Phospholipases Proteins 0.000 description 1
- 102000010780 Platelet-Derived Growth Factor Human genes 0.000 description 1
- 108010038512 Platelet-Derived Growth Factor Proteins 0.000 description 1
- 239000002202 Polyethylene glycol Substances 0.000 description 1
- 108010015078 Pregnancy-Associated alpha 2-Macroglobulins Proteins 0.000 description 1
- 102000006010 Protein Disulfide-Isomerase Human genes 0.000 description 1
- 102000004022 Protein-Tyrosine Kinases Human genes 0.000 description 1
- 108090000412 Protein-Tyrosine Kinases Proteins 0.000 description 1
- 108700020978 Proto-Oncogene Proteins 0.000 description 1
- 102000052575 Proto-Oncogene Human genes 0.000 description 1
- 230000010799 Receptor Interactions Effects 0.000 description 1
- 102100030086 Receptor tyrosine-protein kinase erbB-2 Human genes 0.000 description 1
- 241000283984 Rodentia Species 0.000 description 1
- MTCFGRXMJLQNBG-UHFFFAOYSA-N Serine Natural products OCC(N)C(O)=O MTCFGRXMJLQNBG-UHFFFAOYSA-N 0.000 description 1
- 230000024932 T cell mediated immunity Effects 0.000 description 1
- AYFVYJQAPQTCCC-UHFFFAOYSA-N Threonine Natural products CC(O)C(N)C(O)=O AYFVYJQAPQTCCC-UHFFFAOYSA-N 0.000 description 1
- 239000004473 Threonine Substances 0.000 description 1
- 102000006747 Transforming Growth Factor alpha Human genes 0.000 description 1
- 108090001012 Transforming Growth Factor beta Proteins 0.000 description 1
- 102000004887 Transforming Growth Factor beta Human genes 0.000 description 1
- 101800004564 Transforming growth factor alpha Proteins 0.000 description 1
- 241000209140 Triticum Species 0.000 description 1
- 235000021307 Triticum Nutrition 0.000 description 1
- JXLYSJRDGCGARV-WWYNWVTFSA-N Vinblastine Natural products O=C(O[C@H]1[C@](O)(C(=O)OC)[C@@H]2N(C)c3c(cc(c(OC)c3)[C@]3(C(=O)OC)c4[nH]c5c(c4CCN4C[C@](O)(CC)C[C@H](C3)C4)cccc5)[C@@]32[C@H]2[C@@]1(CC)C=CCN2CC3)C JXLYSJRDGCGARV-WWYNWVTFSA-N 0.000 description 1
- 241000700605 Viruses Species 0.000 description 1
- 230000001594 aberrant effect Effects 0.000 description 1
- 239000000370 acceptor Substances 0.000 description 1
- 230000009858 acid secretion Effects 0.000 description 1
- 230000003213 activating effect Effects 0.000 description 1
- 229960005305 adenosine Drugs 0.000 description 1
- 229960001456 adenosine triphosphate Drugs 0.000 description 1
- 210000001789 adipocyte Anatomy 0.000 description 1
- 239000002671 adjuvant Substances 0.000 description 1
- 239000000695 adrenergic alpha-agonist Substances 0.000 description 1
- 239000000808 adrenergic beta-agonist Substances 0.000 description 1
- 230000002776 aggregation Effects 0.000 description 1
- 238000004220 aggregation Methods 0.000 description 1
- 230000002152 alkylating effect Effects 0.000 description 1
- 238000010171 animal model Methods 0.000 description 1
- 239000005557 antagonist Substances 0.000 description 1
- 229940046836 anti-estrogen Drugs 0.000 description 1
- 230000001833 anti-estrogenic effect Effects 0.000 description 1
- 230000000259 anti-tumor effect Effects 0.000 description 1
- 230000005875 antibody response Effects 0.000 description 1
- 210000000612 antigen-presenting cell Anatomy 0.000 description 1
- 229940065181 bacillus anthracis Drugs 0.000 description 1
- 230000004888 barrier function Effects 0.000 description 1
- WQZGKKKJIJFFOK-VFUOTHLCSA-N beta-D-glucose Chemical compound OC[C@H]1O[C@@H](O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-VFUOTHLCSA-N 0.000 description 1
- 108091008324 binding proteins Proteins 0.000 description 1
- 238000004166 bioassay Methods 0.000 description 1
- 230000008827 biological function Effects 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
- 229960001561 bleomycin Drugs 0.000 description 1
- OYVAGSVQBOHSSS-UAPAGMARSA-O bleomycin A2 Chemical compound N([C@H](C(=O)N[C@H](C)[C@@H](O)[C@H](C)C(=O)N[C@@H]([C@H](O)C)C(=O)NCCC=1SC=C(N=1)C=1SC=C(N=1)C(=O)NCCC[S+](C)C)[C@@H](O[C@H]1[C@H]([C@@H](O)[C@H](O)[C@H](CO)O1)O[C@@H]1[C@H]([C@@H](OC(N)=O)[C@H](O)[C@@H](CO)O1)O)C=1N=CNC=1)C(=O)C1=NC([C@H](CC(N)=O)NC[C@H](N)C(N)=O)=NC(N)=C1C OYVAGSVQBOHSSS-UAPAGMARSA-O 0.000 description 1
- 230000000903 blocking effect Effects 0.000 description 1
- 201000008275 breast carcinoma Diseases 0.000 description 1
- 230000003491 cAMP production Effects 0.000 description 1
- 239000012830 cancer therapeutic Substances 0.000 description 1
- 231100000504 carcinogenesis Toxicity 0.000 description 1
- 230000000747 cardiac effect Effects 0.000 description 1
- 210000004413 cardiac myocyte Anatomy 0.000 description 1
- 239000000969 carrier Substances 0.000 description 1
- 150000001768 cations Chemical class 0.000 description 1
- 230000021164 cell adhesion Effects 0.000 description 1
- 210000002421 cell wall Anatomy 0.000 description 1
- 239000013000 chemical inhibitor Substances 0.000 description 1
- 239000003153 chemical reaction reagent Substances 0.000 description 1
- 229940044683 chemotherapy drug Drugs 0.000 description 1
- 229960001231 choline Drugs 0.000 description 1
- OEYIOHPDSNJKLS-UHFFFAOYSA-N choline Chemical compound C[N+](C)(C)CCO OEYIOHPDSNJKLS-UHFFFAOYSA-N 0.000 description 1
- 230000001684 chronic effect Effects 0.000 description 1
- 230000004087 circulation Effects 0.000 description 1
- OHCQJHSOBUTRHG-UHFFFAOYSA-N colforsin Natural products OC12C(=O)CC(C)(C=C)OC1(C)C(OC(=O)C)C(O)C1C2(C)C(O)CCC1(C)C OHCQJHSOBUTRHG-UHFFFAOYSA-N 0.000 description 1
- 210000004922 colonic epithelial cell Anatomy 0.000 description 1
- 230000002281 colonystimulating effect Effects 0.000 description 1
- 230000000052 comparative effect Effects 0.000 description 1
- 238000006482 condensation reaction Methods 0.000 description 1
- 230000001268 conjugating effect Effects 0.000 description 1
- 230000008602 contraction Effects 0.000 description 1
- IDLFZVILOHSSID-OVLDLUHVSA-N corticotropin Chemical compound C([C@@H](C(=O)N[C@@H](CO)C(=O)N[C@@H](CCSC)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC=1NC=NC=1)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)NCC(=O)N[C@@H](CCCCN)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](C(C)C)C(=O)NCC(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H](CC=1C=CC(O)=CC=1)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CC(N)=O)C(=O)NCC(=O)N[C@@H](C)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CO)C(=O)N[C@@H](C)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](C)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC=1C=CC=CC=1)C(O)=O)NC(=O)[C@@H](N)CO)C1=CC=C(O)C=C1 IDLFZVILOHSSID-OVLDLUHVSA-N 0.000 description 1
- 229960000258 corticotropin Drugs 0.000 description 1
- 235000018417 cysteine Nutrition 0.000 description 1
- XUJNEKJLAYXESH-UHFFFAOYSA-N cysteine Natural products SCC(N)C(O)=O XUJNEKJLAYXESH-UHFFFAOYSA-N 0.000 description 1
- 210000004292 cytoskeleton Anatomy 0.000 description 1
- 231100000433 cytotoxic Toxicity 0.000 description 1
- 239000002254 cytotoxic agent Substances 0.000 description 1
- 229940127089 cytotoxic agent Drugs 0.000 description 1
- 231100000599 cytotoxic agent Toxicity 0.000 description 1
- 238000011393 cytotoxic chemotherapy Methods 0.000 description 1
- 230000001472 cytotoxic effect Effects 0.000 description 1
- 231100000135 cytotoxicity Toxicity 0.000 description 1
- 230000003013 cytotoxicity Effects 0.000 description 1
- 230000007812 deficiency Effects 0.000 description 1
- 230000003413 degradative effect Effects 0.000 description 1
- 230000002939 deleterious effect Effects 0.000 description 1
- 238000001212 derivatisation Methods 0.000 description 1
- 238000011161 development Methods 0.000 description 1
- 125000002228 disulfide group Chemical group 0.000 description 1
- 238000009509 drug development Methods 0.000 description 1
- 230000003028 elevating effect Effects 0.000 description 1
- 210000002889 endothelial cell Anatomy 0.000 description 1
- 210000003038 endothelium Anatomy 0.000 description 1
- 239000002158 endotoxin Substances 0.000 description 1
- 230000009088 enzymatic function Effects 0.000 description 1
- 229940105423 erythropoietin Drugs 0.000 description 1
- 229960005309 estradiol Drugs 0.000 description 1
- 229930182833 estradiol Natural products 0.000 description 1
- 229940011871 estrogen Drugs 0.000 description 1
- 239000000262 estrogen Substances 0.000 description 1
- 239000000328 estrogen antagonist Substances 0.000 description 1
- 230000028023 exocytosis Effects 0.000 description 1
- 102100021145 fMet-Leu-Phe receptor Human genes 0.000 description 1
- 239000003527 fibrinolytic agent Substances 0.000 description 1
- 230000003480 fibrinolytic effect Effects 0.000 description 1
- 210000002950 fibroblast Anatomy 0.000 description 1
- 239000000945 filler Substances 0.000 description 1
- 238000001914 filtration Methods 0.000 description 1
- 229940028334 follicle stimulating hormone Drugs 0.000 description 1
- 239000012634 fragment Substances 0.000 description 1
- 229940044627 gamma-interferon Drugs 0.000 description 1
- 230000002496 gastric effect Effects 0.000 description 1
- 238000002523 gelfiltration Methods 0.000 description 1
- 102000034356 gene-regulatory proteins Human genes 0.000 description 1
- 108091006104 gene-regulatory proteins Proteins 0.000 description 1
- 102000054766 genetic haplotypes Human genes 0.000 description 1
- 210000004602 germ cell Anatomy 0.000 description 1
- 230000001434 glomerular Effects 0.000 description 1
- 239000008103 glucose Substances 0.000 description 1
- 230000006377 glucose transport Effects 0.000 description 1
- 229960003180 glutathione Drugs 0.000 description 1
- 230000013595 glycosylation Effects 0.000 description 1
- 238000006206 glycosylation reaction Methods 0.000 description 1
- 125000003630 glycyl group Chemical group [H]N([H])C([H])([H])C(*)=O 0.000 description 1
- 210000000224 granular leucocyte Anatomy 0.000 description 1
- 239000000122 growth hormone Substances 0.000 description 1
- 230000003394 haemopoietic effect Effects 0.000 description 1
- 125000005842 heteroatom Chemical group 0.000 description 1
- 231100000086 high toxicity Toxicity 0.000 description 1
- 210000003630 histaminocyte Anatomy 0.000 description 1
- 125000000487 histidyl group Chemical group [H]N([H])C(C(=O)O*)C([H])([H])C1=C([H])N([H])C([H])=N1 0.000 description 1
- BIHNZKYXOTXXSP-GCBHNEMQSA-N holotoxin a Chemical compound O[C@@H]1[C@@H](OC)[C@H](O)[C@@H](CO)O[C@H]1O[C@@H]1[C@@H](O)[C@H](O[C@H]2[C@@H]([C@@H](O[C@H]3[C@@H]([C@@H](O)[C@H](O[C@H]4[C@@H]([C@@H](O[C@H]5[C@@H]([C@@H](OC)[C@H](O)[C@@H](CO)O5)O)[C@H](O)[C@@H](CO)O4)O)[C@@H](C)O3)O)[C@H](OC3C(C4[C@](C=5C([C@@]6(CC(=O)[C@H]7C6(C(O[C@]7(CCCC(C)=C)C)=O)CC=5)C)CC4)(C)CC3)(C)C)OC2)O)O[C@H](CO)[C@H]1O BIHNZKYXOTXXSP-GCBHNEMQSA-N 0.000 description 1
- 230000028996 humoral immune response Effects 0.000 description 1
- 230000008348 humoral response Effects 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
- 238000003384 imaging method Methods 0.000 description 1
- 229940127121 immunoconjugate Drugs 0.000 description 1
- 230000001506 immunosuppresive effect Effects 0.000 description 1
- 238000009169 immunotherapy Methods 0.000 description 1
- 230000000415 inactivating effect Effects 0.000 description 1
- 238000010348 incorporation Methods 0.000 description 1
- 230000002757 inflammatory effect Effects 0.000 description 1
- 230000004054 inflammatory process Effects 0.000 description 1
- 238000001802 infusion Methods 0.000 description 1
- 238000003780 insertion Methods 0.000 description 1
- 230000037431 insertion Effects 0.000 description 1
- 229940079322 interferon Drugs 0.000 description 1
- 230000018276 interleukin-1 production Effects 0.000 description 1
- 229940047122 interleukins Drugs 0.000 description 1
- 230000037041 intracellular level Effects 0.000 description 1
- 230000004068 intracellular signaling Effects 0.000 description 1
- 230000007728 intracellular signaling mechanism Effects 0.000 description 1
- 238000007918 intramuscular administration Methods 0.000 description 1
- 238000007912 intraperitoneal administration Methods 0.000 description 1
- 230000002427 irreversible effect Effects 0.000 description 1
- 210000004153 islets of langerhan Anatomy 0.000 description 1
- 239000000644 isotonic solution Substances 0.000 description 1
- 239000002523 lectin Substances 0.000 description 1
- 231100001231 less toxic Toxicity 0.000 description 1
- 208000032839 leukemia Diseases 0.000 description 1
- YEESKJGWJFYOOK-IJHYULJSSA-N leukotriene D4 Chemical compound CCCCC\C=C/C\C=C/C=C/C=C/[C@H]([C@@H](O)CCCC(O)=O)SC[C@H](N)C(=O)NCC(O)=O YEESKJGWJFYOOK-IJHYULJSSA-N 0.000 description 1
- 230000004130 lipolysis Effects 0.000 description 1
- 230000007774 longterm Effects 0.000 description 1
- 229940040129 luteinizing hormone Drugs 0.000 description 1
- 210000003794 male germ cell Anatomy 0.000 description 1
- 125000005439 maleimidyl group Chemical group C1(C=CC(N1*)=O)=O 0.000 description 1
- 210000004962 mammalian cell Anatomy 0.000 description 1
- 239000000463 material Substances 0.000 description 1
- 201000001441 melanoma Diseases 0.000 description 1
- 102000006240 membrane receptors Human genes 0.000 description 1
- 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 1
- 230000005012 migration Effects 0.000 description 1
- 238000013508 migration Methods 0.000 description 1
- 230000002297 mitogenic effect Effects 0.000 description 1
- 238000006011 modification reaction Methods 0.000 description 1
- 108091005573 modified proteins Proteins 0.000 description 1
- 102000035118 modified proteins Human genes 0.000 description 1
- 239000003607 modifier Substances 0.000 description 1
- 230000009456 molecular mechanism Effects 0.000 description 1
- 210000000865 mononuclear phagocyte system Anatomy 0.000 description 1
- 230000003551 muscarinic effect Effects 0.000 description 1
- 210000003205 muscle Anatomy 0.000 description 1
- 210000000822 natural killer cell Anatomy 0.000 description 1
- 230000010309 neoplastic transformation Effects 0.000 description 1
- PCJGZPGTCUMMOT-ISULXFBGSA-N neurotensin Chemical compound C([C@@H](C(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@@H](CC(C)C)C(O)=O)NC(=O)[C@H]1N(CCC1)C(=O)[C@H](CCCN=C(N)N)NC(=O)[C@H](CCCN=C(N)N)NC(=O)[C@H]1N(CCC1)C(=O)[C@H](CCCCN)NC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)[C@H](CC(C)C)NC(=O)[C@H]1NC(=O)CC1)C1=CC=C(O)C=C1 PCJGZPGTCUMMOT-ISULXFBGSA-N 0.000 description 1
- 230000007135 neurotoxicity Effects 0.000 description 1
- 231100000228 neurotoxicity Toxicity 0.000 description 1
- 230000007935 neutral effect Effects 0.000 description 1
- 230000003472 neutralizing effect Effects 0.000 description 1
- 230000002560 nonimmunologic effect Effects 0.000 description 1
- 239000002773 nucleotide Substances 0.000 description 1
- 125000003729 nucleotide group Chemical group 0.000 description 1
- 108091008819 oncoproteins Proteins 0.000 description 1
- 230000001936 parietal effect Effects 0.000 description 1
- 239000000813 peptide hormone Substances 0.000 description 1
- 210000003819 peripheral blood mononuclear cell Anatomy 0.000 description 1
- 230000002093 peripheral effect Effects 0.000 description 1
- 230000008782 phagocytosis Effects 0.000 description 1
- 239000008177 pharmaceutical agent Substances 0.000 description 1
- 239000000825 pharmaceutical preparation Substances 0.000 description 1
- 229940127557 pharmaceutical product Drugs 0.000 description 1
- 230000000144 pharmacologic effect Effects 0.000 description 1
- SONNWYBIRXJNDC-VIFPVBQESA-N phenylephrine Chemical compound CNC[C@H](O)C1=CC=CC(O)=C1 SONNWYBIRXJNDC-VIFPVBQESA-N 0.000 description 1
- 229960001802 phenylephrine Drugs 0.000 description 1
- 239000010452 phosphate Substances 0.000 description 1
- 239000008363 phosphate buffer Substances 0.000 description 1
- 229910052697 platinum Inorganic materials 0.000 description 1
- BASFCYQUMIYNBI-UHFFFAOYSA-N platinum Substances [Pt] BASFCYQUMIYNBI-UHFFFAOYSA-N 0.000 description 1
- 230000004983 pleiotropic effect Effects 0.000 description 1
- 229920001223 polyethylene glycol Polymers 0.000 description 1
- 229920000642 polymer Polymers 0.000 description 1
- 229920001184 polypeptide Polymers 0.000 description 1
- 238000010837 poor prognosis Methods 0.000 description 1
- 230000004481 post-translational protein modification Effects 0.000 description 1
- 230000003389 potentiating effect Effects 0.000 description 1
- 230000002028 premature Effects 0.000 description 1
- 238000002360 preparation method Methods 0.000 description 1
- 125000002924 primary amino group Chemical group [H]N([H])* 0.000 description 1
- 230000037452 priming Effects 0.000 description 1
- 230000009696 proliferative response Effects 0.000 description 1
- 108020003519 protein disulfide isomerase Proteins 0.000 description 1
- 108020001580 protein domains Proteins 0.000 description 1
- 230000002285 radioactive effect Effects 0.000 description 1
- 238000001959 radiotherapy Methods 0.000 description 1
- 239000000376 reactant Substances 0.000 description 1
- 238000009877 rendering Methods 0.000 description 1
- 230000003252 repetitive effect Effects 0.000 description 1
- 230000000717 retained effect Effects 0.000 description 1
- 238000012552 review Methods 0.000 description 1
- 206010039073 rheumatoid arthritis Diseases 0.000 description 1
- 210000003705 ribosome Anatomy 0.000 description 1
- 238000000926 separation method Methods 0.000 description 1
- 150000003384 small molecules Chemical class 0.000 description 1
- 230000016160 smooth muscle contraction Effects 0.000 description 1
- 239000001488 sodium phosphate Substances 0.000 description 1
- 229910000162 sodium phosphate Inorganic materials 0.000 description 1
- 239000012064 sodium phosphate buffer Substances 0.000 description 1
- 239000007790 solid phase Substances 0.000 description 1
- 125000006850 spacer group Chemical group 0.000 description 1
- 241000894007 species Species 0.000 description 1
- 238000001228 spectrum Methods 0.000 description 1
- 210000000952 spleen Anatomy 0.000 description 1
- 238000003860 storage Methods 0.000 description 1
- 238000007920 subcutaneous administration Methods 0.000 description 1
- 230000001839 systemic circulation Effects 0.000 description 1
- 150000003568 thioethers Chemical class 0.000 description 1
- 229940104230 thymidine Drugs 0.000 description 1
- 210000001541 thymus gland Anatomy 0.000 description 1
- 231100000155 toxicity by organ Toxicity 0.000 description 1
- 230000007675 toxicity by organ Effects 0.000 description 1
- 238000012546 transfer Methods 0.000 description 1
- 238000002054 transplantation Methods 0.000 description 1
- RYFMWSXOAZQYPI-UHFFFAOYSA-K trisodium phosphate Chemical compound [Na+].[Na+].[Na+].[O-]P([O-])([O-])=O RYFMWSXOAZQYPI-UHFFFAOYSA-K 0.000 description 1
- OUYCCCASQSFEME-UHFFFAOYSA-N tyrosine Natural products OC(=O)C(N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-UHFFFAOYSA-N 0.000 description 1
- 230000004222 uncontrolled growth Effects 0.000 description 1
- 210000002700 urine Anatomy 0.000 description 1
- 230000024883 vasodilation Effects 0.000 description 1
- 230000002861 ventricular Effects 0.000 description 1
- 230000035899 viability Effects 0.000 description 1
- 229960003048 vinblastine Drugs 0.000 description 1
- JXLYSJRDGCGARV-XQKSVPLYSA-N vincaleukoblastine Chemical compound C([C@@H](C[C@]1(C(=O)OC)C=2C(=CC3=C([C@]45[C@H]([C@@]([C@H](OC(C)=O)[C@]6(CC)C=CCN([C@H]56)CC4)(O)C(=O)OC)N3C)C=2)OC)C[C@@](C2)(O)CC)N2CCC2=C1NC1=CC=CC=C21 JXLYSJRDGCGARV-XQKSVPLYSA-N 0.000 description 1
- 230000029812 viral genome replication Effects 0.000 description 1
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 1
- 239000003643 water by type Substances 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/50—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
- A61K47/51—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
- A61K47/62—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
- A61K47/64—Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
- A61K47/6425—Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent the peptide or protein in the drug conjugate being a receptor, e.g. CD4, a cell surface antigen, i.e. not a peptide ligand targeting the antigen, or a cell surface determinant, i.e. a part of the surface of a cell
Definitions
- the present invention relates to therapeutic conjugates useful in activating, suppressing, or in some way altering a biological response in target cells.
- Such conjugates include a proteinaceous non- cytolytic toxin moiety conjugated by chemical or recombinant means to a targeting moiety.
- BRMs proteinaceous biological response modifiers
- IL- 1 Macrophages activated Most endstage and T- cells . Many other progenitor lymphoid cells. lymphoid and non- lymphoid cells .
- IL- 2 Activated T-cells .
- IL-3 T-cell ⁇ Early progenitor cells, mast cells.
- T-cells Activated T-cells .
- T-cell ⁇ B-cells.
- I -5 T-cells .
- B-cells eo ⁇ inophils.
- I - 6 Macrophages activated B-cells, hepatocytes.
- T-cells and other non- lymphoid cells .
- T- 7 Activated T-cells .
- IL- 8 Activated T-cells, PMN ⁇ . Endothelial cells.
- G -CSF Activated T-cells. Granulocytic and myeloid end ⁇ tage and progenitor cells, T-cells , monocytes .
- G-CSF Activated T-cells Granulocytic endstage and progenitor cells .
- M-CSF Macrophages Monocytic endstage and or progenitor cells .
- BRMs has grown, common elements in the molecular mechanisms of action and in the regulation of BRMs as well as evolutionary relationships between the varied BRMs have become apparent .
- the mechanism of action of BRMs varies according to the particular BRM and target cell involved.
- studies with GM- CSF indicate that a second messenger, cAMP, is involved in mediating the priming of monocytes for enhanced antigen presentation.
- cAMP a second messenger
- no role for cAMP has been demonstrated in GM-CSF activation of polymorphonuclear leukocytes for oxidative burst. See Di Persio et al. , J. Immunol.. 140:4315. 1988.
- one BRM-induced activity may be mediated through an effector molecule such as tyrosine kinase while another BRM-induced activity occurring within the same target cell may not.
- This phenomenon was evidenced with IL-2 in Mills et al., Cell. 55.:91, 1988.
- the key to the induction of a target cell response to a given BRM appears to be the presence of a specific receptor on the target cell.
- BRM activity is regulated in a variety of ways. Both positive and negative regulators of BRM activity exist. Molecules such as GM-CSF, G-CSF, IL-3 and EPO that stimulate proliferation of bone marrow progenitor cells have been known for some time, but inhibitors of proliferation also exist.
- Such proliferation inhibitors are induced upon binding of the positive regulator (e.g.. G-CSF) and lead to a dampening of the positive signal (i.e. , proliferation) and the return of progenitor cells to the quiescent state.
- the positive regulator e.g. G-CSF
- Such inhibitors may also be produced constititively in low amounts and participate in maintaining only a small proportion of progenitor cells in an active state of proliferation.
- Duration of target cell response is dependent upon a number of factors including the serum half- life of the response-inducing BRM. See Cheever et al., . Immunol.. 134.:3895, 1985 and Metcalf et al., Proc. Nat'l Acad. Sci. (USA). £5.:3160, 1988. Conse- quently, serum half-life can be modulated to increase in vivo BRM activity. BRMs with longer serum half- lives are being developed for therapeutic applications to reduce the requirement for continuous infusion of the BRM and to prolong BRM activity in vivo. See Knauf et al. , J. Biol. Chem.. 161:15064, 1988 and Takakura et al., Pharm. Res.
- Serum half-life of a BRM is a function of a number of factors, such as 1) receptor- mediated uptake of BRMs, usually evidenced by accumulation in hematopoietic or RES (i.e.. reticuloendothelial system) organs; 2) non-specific uptake of BRMs, a function of BRM hydrophobicity and charge; 3) size of BRMs, since proteins larger than albumin generally have longer serum half-lives than proteins smaller than albumin due to lower rates of kidney filtration and excretion; 4) rate of metabolism and excretion of BRMs; and 5) binding or interaction of BRMs with serum glycoproteins or soluble receptors.
- RES i.e.. reticuloendothelial system
- BRMs modulate the migration of leukocytes to sites of inflammation or leukocyte egress into the circulation.
- the arrival of leukocytes at inflammatory sites provides additional cellular sources for cytokine BRM stimulation and amplification. See Wang et al., Blood. 72:1456, 1988.
- eukaryotic cell BRM activity appears to be mediated through cellular receptors.
- These receptors may vary in size, glycosylation, or surface expression, possibly even reflecting individual cellular differences in post- translational modification of the same gene product.
- CSF colony stimulating factor
- Comparative studies with characterized CSF (i.e.. colony stimulating factor) receptors from different cell types indicate that the receptor may differ biochemically from cell type to cell type (Baldwin et al., Blood, 71:1033, 1989) even though these receptors may be related evolutionarily. See Durum et al., Immunol. Today, .11:103, 1990. Biochemical differences in receptors may lead to functional changes in receptor expression or affinity.
- BRM binding or uptake into tissues need not lead to any significant response. This has been recognized from bio-distribution studies with labeled eukaryotic BRMs. BRMs typically show high uptake into RES organs such as the liver and spleen as well as uptake into tissues not associated with a biological response. This latter uptake may reflect binding to non-functional receptors or non-specific uptake. In addition, tissues to which BRMs bind that are not associated with a BRM response may well exhibit such a response, with the cellular activity escaping the notice of investigators because that activity is not of sufficient magnitude or being investigated.
- BRMs also potentially modulating cellular response to BRMs is the interaction of those BRMs with soluble serum receptors, glycoproteins, or antibodies.
- Soluble receptors or binding proteins have been characterized from human urine or serum for a variety of cytokines. It is likely therefore that most cytokines undergo some interaction with serum constituents.
- Alpha-2-macroglobulin is a serum protein known to interact with multiple cytokines. The effect of this serum protein-BRM interaction is unknown, but such interactions may serve to prolong the serum half-life of BRMs.
- Recent findings also indicate that antibodies to cytokines exist in normal sera.
- Cytokine synthesis for example, has been shown to be induced in a cascading fashion after binding of cytokines to their receptors, probably through activation of oncogenes and DNA binding proteins or through activation of second messengers. See Farrar et al., CRC Critical Reviews in Therapeutic Drucr Carrier Systems. 1:229, 1989.
- BRM- stimulated IL-1 secretion by monocytes leads to the induction of tandemly regulated genes and coordinated synthesis of IL-2, IL-4, and GM-CSF in T-cells. See Oppenheim et al., Immunol. Letters, 16 . :179, 1987.
- Mitogenic stimulation of T-cells can lead to coordinated synthesis of TNF and gamma-interferon.
- a given biological response can be directly attributed to binding of a cytokine to its receptor on the target cell or can arise indirectly through cytokine interaction with another cell type that then secretes or expresses one or more lymphokines, inducing a pleiotropic target cell or tissue response. Consequently, some of the biological activities of IL-2 and its usefulness in cancer therapy may result from its direct stimulation of sensitized T-cells (Mule et al., . Immunol.. 139:285, 1987) while much of its associated toxicity may result from its induction of TNF which leads to secondary toxicities. See Mier et al., J. Clin. Immunol.. 8.:426, 1988.
- BRM activity has also been associated with bacterial cell products.
- IL-1 has a spectrum of biological activities very similar to endotoxin, a cell wall constituent of gram negative bacteria.
- a number of bacterial compounds have been shown to have multiple BRM activities, including colony stimulating and IL-2- inducing activity. These activities have usually been associated with crude bacterial preparations, however.
- bacterial enterotoxins or exotoxins are also a rich source of BRM activity.
- Bacterial toxin BRMs share certain characteristics with their eukaryotic cell BRM counterparts. Both types of BRMs are soluble molecules and primarily induce effects in the region of the target cell but are capable of producing more distant effects. BRMs from bacterial toxins, however, differ markedly from eukaryotic BRMs in that the bacterial toxin BRMs exhibit an enzymatic (i.e. , toxic) activity associated with a particular structural domain or subunit.
- BRMs may be useful in a wide variety of therapeutic applications.
- the ability of BRMs to impact many aspects of the immune system make such molecules attractive in the treatment of cancer, autoimmune deficiencies, and other conditions impacting the immune system.
- a major goal for cancer therapy has been to improve the selectivity of otherwise toxic therapies by taking advantage of certain attributes of tumor cells that are distinctive from most normal cells.
- the degree of selectivity of a therapeutic agent is usually assessed by its therapeutic index, i.e.. the ratio of the potency of the therapeutic agent against a tumor to the toxicity of the agent with respect to normal cells. Consequently, cancer therapy can be improved by one or more of the following: (1) increasing the potency of the therapeutic agent against the tumor while not changing toxicity to normal organs; (2) decreasing the toxicity of the therapeutic agent to dose limiting normal organs; (3) improving the selectivity of the therapeutic agent for tumor cells over normal cells by targeting of the cytotoxic agent; or (4) delivering a molecule having effects on tumor cells (e.g..).
- Protein toxins for example, have been employed as toxic moieties in prior art conjugates referred to as immunotoxins. All of the toxins heretofore used in immunotoxin construction affect protein synthesis within the cell. Intact toxins or toxin portions used heretofore include diphtheria, Pseudomonas exotoxin A, shigella, ricin and abrin (plant) . Plant toxins also occur as enzymatic subunits devoid of a cell binding domain, namely ribosomal inactivating proteins (RIPs) and include pokeweed anti-viral protein, gelonin, modecin, and an inhibitor from barley or wheat germ and the like.
- RIPs ribosomal inactivating proteins
- toxins Since protein synthesis is an essential cellular function, these toxins should have the ability to kill any cell to which the toxins are targeted, including cells normally resistant to chemotherapy or radiation treatment.
- the degree of selectivity in target cell killing of an immunotoxin is a function of the type of toxin, whether the whole toxin or only the portion of the toxin responsible for enzymatic activity is conjugated to the targeting moiety, the bond(s) used to conjugate the toxin to the targeting moiety, and factors affecting in vivo delivery. See Morgan et al. , Antibody Immunocon ucrates and Radio Pharmaceuticals. 2.:165, 1989; and Morgan et al., Mol. Immunol. , 22:273, 1990.
- Toxins employed in immunotoxins as described above exhibit high toxicity, at least in holotoxin form, and high immunogenicity. Specifically, toxins adversely affect normal cells through receptor- mediated or non-specific uptake and, as a result, efficacy of the toxin against target cells is limited. Because these toxins inhibit an essential cellular function, e.g.. protein synthesis, their non-target tissue uptake results in normal organ toxicity.
- toxin also generates a strong immune response in vivo.
- the recipient's immune system recognizes the d_ ⁇ munotoxin and removes it from the systemic circulation, thereby reducing the immunotoxin therapeutic efficacy.
- immunotoxins are expected to exhibit no selectivity for metabolic processes within tumor cells compared to normal tissues. Target cell metabolism may also be manipulated to selectively achieve a therapeutic goal, however. An example of such selectivity is evidenced by the non-targeted agents affecting cAMP dependent protein kinase and cAMP levels.
- PKA cAMP dependent protein kinase
- 8-Cl-cAMP An example of a moiety capable of affecting cAMP dependent protein kinase (PKA) is 8-Cl-cAMP.
- the agent binds irreversibly to PKA, thereby halting proliferation of target cells which is dependent upon the enzyme.
- This agent has been demonstrated to inhibit proliferation of a variety of human and rodent tumor cell lines.
- Drug development aimed at identifying and/or designing drugs capable of discrimination between altered or amplified forms of protein kinase present in tumor tissues and in normal tissues is currently being pursued.
- anti-sense oligonucleotides to PKA have recently been shown to inhibit proliferation of tumor cells.
- G-proteins are signaling moieties acting between certain receptors and effector molecules within cells. Ligand binding induces a signal which is communicated to effector molecules through modification and then subsequent interaction with the G-protein alpha subunits. The activated G-protein signals that the effector moiety should either commence or halt its effector function.
- effector molecules i.e.. enzymatic or ion channel proteins, mediate changes in cellular processes.
- the receptors, G- proteins, and effector molecules are located in proximity to each other within the cell plasma membrane (Fig. 1) .
- G-proteins are typically present as three-subunits, alpha, beta, and gamma. Effector molecules are generally capable of freely diffusing within the plasma membrane or cytoplasm and couple with an activated G-alpha subunit.
- the appropriate G-alpha subunit When a ligand binds to its receptor as shown in Fig. 1, the appropriate G-alpha subunit is activated by the exchange of GTP for GDP.
- the activated G- alpha subunit which is released from association with the beta and gamma subunits, then interacts with an effector moiety, such as adenyl cyclase, to activate or deactivate the effector moiety.
- an effector moiety such as adenyl cyclase
- Activation of the effector adenyl cyclase results in the cleavage of ATP to form cAMP.
- the receptor becomes unoccupied, i.e.. the ligand-receptor interaction is lost, the G-alpha subunit will be de-activated and reassociate with the other G-protein subunits. Consequently, a loss of signal transduction will be experienced, and cAMP levels will return to normal.
- the ligand will be bound to the receptor for only a short time. Conse ⁇ quently, the biological response induced by the receptor-ligand interaction is typically of short duration and is dictated by the concentration of ligand in proximity to a receptor.
- pertussis and cholera toxin A large variety of cellular responses have been shown to be mediated through G-proteins or changes in cyclic nucleotide levels by the use of specific bacterial toxins, namely pertussis and cholera. These toxins interact with different G-alpha proteins to uncouple receptor signaling of cAMP production (inhibitory) or to prolong the activated state of the G-alpha s subunit (stimulatory) , thereby increasing cellular levels of cAMP. Thus, in this sense, pertussis and cholera toxin may be viewed as the "yin and yang" for G-proteins. Using those toxins as analytical tools, investigators have examined specific target cells and receptor-ligand interactions to ascertain whether a given biological response is mediated through G-proteins.
- Target cells "T” are either pre-treated with toxin or simultaneously treated with ligand L and holotoxin "A/B.”
- Holotoxin A/B binds through its B subunit to cellular acceptors, internalizes and catalyzes ADP- ribosylation of G-proteins thereby affecting signaling.
- a diminution or enhancement of the biological response triggered by ligand L binding to receptor "R” is taken as evidence of receptor- effector coupling through G-proteins.
- Tables III and IV Effector moieties coupled to receptors through G-proteins are regulatable in accordance with the present invention.
- Exemplary effector moieties linked to receptors through G-protein substrates are adenyl cyclase, phospholipase, phosphodiesterase, ion channels, cAMP dependent protein kinase, and the like.
- the functioning of these exemplary effector moieties may be impacted by the toxin portion of conjugates of the present invention, operating through existing target cell signaling mechanisms.
- Table III lists exemplary immunologic responses induced by ligand-receptor interactions and regulated by cyclic nucleotides or G-proteins. TABLE III
- Hepatocytes Hepatocytes, CI-inhibitor Histamine, PGE- Monocytes, 2 , C5a des arg, Fibroblasts, Carba yl Endothelium choline, Phenylephrine , Interferon ⁇
- Table IV lists exemplary non- immuno logic responses induced by ligand- receptor interactions and regulated by cyclic nucleotides or G-proteins .
- Adipocytes Glucose Transport Insulin Lipolysis Pertussis Toxin
- Ca ++ dependent ATPase Calmodulin Pharmaceutical agents that can modulate the variety of biological responses listed in these tables may find significant utility. If such agents include an appropriate holotoxin, they would have the inherent advantage of high potency due to the enzymatic nature of the toxin. However, if advantage is to be taken of the ability of a holotoxin to diminish or amplify an in vivo biological response triggered by ligand binding, several obstacles must be overcome. First, the holotoxin is toxic to certain normal cells, limiting the toxin dose and therefore the amount of signal regulation achievable in target cells bound by the ligand. In addition, the holotoxin is not selective for receptor-positive target cells.
- One method of curing a mammal of a tumor is to kill the tumor cells while preserving the viability of the host. This is generally the approach of radiation therapy, chemotherapy and immunotherapy.
- Another approach is to cause the tumor cells to differentiate, since the differentiated form of a cell is an end-stage cell.
- anecdotes of cis-retinoic acid causing differentiation of human leukemia.
- chemotherapy may partially kill and partially cause differentiation into benign tumors.
- tissue culture nontoxic levels of cAMP in the media down-regulates certain cellular functions.
- the present invention provides conjugates that retarget non- cytolytic toxin moieties to amplify or suppress differentiation functions of target cells.
- Conjugates of the first aspect of the present invention include an enzymatic portion of a non- cytolytic toxin capable of ADP-ribosylation of a G-protein and a targeting agent (ligand) capable of binding to a target cell receptor, which is coupled through G-proteins to effector molecules capable of impacting the desired biological response.
- the receptor is at least somewhat specific to the target cells so that the ligand-BRM conjugate is at least preferentially delivered to those cells, such as tumor cells, for which manipulation is desired.
- Toxin moieties and subunits derived therefrom useful in this aspect of the present invention include toxins that do not affect protein synthesis directly but rather interact with G-proteins or modified G-proteins (oncogenes ) .
- Exemplary toxins from which enzymatic moieties can be derived are listed in Table V.
- E . coli heat GM-1, GDlb, Gs alpha labile enterotoxin a ⁇ ialo-GM-1
- the conjugates of the present invention include toxin moieties having their endogenous cell binding capability nullified or removed by biochemical or recombinant genetic engineering methods or by occlusion occurring upon conjugation to the targeting agent. In this manner, the conjugate is retargeted in accordance with the cell binding specificity of the targeting moiety, rather than the endogenous cell binding activity of the toxin. In all instances, the final conjugate retains the enzymatic activity of the holotoxin.
- Such retargeted conjugates are less toxic than the native holotoxin, because the conjugated toxin moiety no longer exhibits the endogenous cell binding specificity which elicits the normal dose-limiting toxicity pattern of the holotoxin. Moreover, targeting G-proteins and the cellular processes regulated by these proteins will not lead to cellular death in most cases but merely to an alteration of a particular biological response. Target cell selectivity (i.e.. bioavailability of the conjugate for target cells) is also improved by retargeting.
- Retargeting non-cytolytic toxins using a ligand specific for a receptor that is preferentially expressed on target cells will also impart a longer serum half life and higher bioavailability than holotoxin, as the receptors, for instance, for pertussis and cholera, are widely displayed over many cell types.
- ADP-ribosylation of G-proteins by exemplary non-cytolytic toxins cholera and pertussis is essentially irreversible and, as a result, the effects on the biological response are of extended duration with the effected cell recovering normal signal regulation only through synthesis of new G- proteins. Consequently, conjugates of the present invention employing portions of non-cytolytic toxins will be characterized by achieving a prolonged effect on the targeted biological response, with normal biological response recovery dictated by the rate of target cell G-protein synthesis.
- a second aspect of the present invention provides conjugates capable of affecting functions of target cells independently of target cell signaling mechanisms and G-proteins.
- Such conjugates include a non-cytolytic toxin moiety capable of directly catalyzing an intracellular reaction that affects target cell function and a targeting moiety.
- Exemplary holotoxins useful in a direct-acting conjugate embodiment of the present invention are adenylate cyclase toxins. Such toxins catalyze the conversion of ATP to cAMP and are functionally equivalent to mammalian adenyl cyclase.
- these toxins do not operate through a cellular signaling system, the enzymatic portion of these direct-acting toxins may be retargeted by any moiety selective for the target cells, without regard to whether the receptor for that moiety is linked to G-proteins.
- metabolic manipulation can be achieved in target cells, such as tumor cells, that may be abnormal in their signal transduction.
- adenyl cyclase toxins from which the toxin moieties employed in conjugates of this embodiment of the present invention are derived, are active for only a short time in target cells as a result of host cell enzymatic degradation of the toxin.
- Conjugates of this and other aspects of the present invention may therefore include toxin moieties modified to be less susceptible to degradation or inactivation, thereby maintaining elevated cAMP levels and altered biological responses for prolonged time periods.
- a third aspect of the present invention is provided wherein a moiety derived from a non- cytolytic toxin capable of ADP-ribosylation of actin is conjugated to and retargeted by a targeting agent such as ligand or antibody.
- a targeting agent such as ligand or antibody.
- Table VI lists exemplary toxins useful in the second and third aspects of the present invention. TABLE VI
- Conjugates of the present invention may also be manipulated by a number of methods to reduce the immunogenicity thereof, a problem inherent in the development of pharmaceutical products based on toxins.
- Fig. 1 is a schematic representation of how G- proteins couple ligand-receptor binding to effector functions within cells.
- Fig. 2a is a schematic representation of experiments performed to determine whether a given target cell receptor-ligand binding and a subsequent biologic response are coupled through G-proteins. The determination is made by pretreating or simultaneously treating a cell with a toxin that modifies G-protein activity in conjunction with ligand-receptor interaction.
- Fig. 2b is a schematic representation of the retargeted, non-cytolytic toxin conjugates of the first aspect of the present invention.
- Fig. 3 is a schematic representation of the structure of cholera holotoxin.
- Fig. 4 is a schematic representation of a retargeted cholera toxin conjugate of the first aspect of the present invention.
- Fig. 5 is a schematic representation of the structure of adenylate cyclase toxin (ACT) of Bordetella pertussis.
- ACT adenylate cyclase toxin
- Fig. 6 is a schematic representation of recombinant derivatives of ACT useful in retargeted toxin conjugates of the second aspect of the present invention.
- Fig. 7 is a schematic representation of a conditionally unstable peptide linkage useful in optimizing potency and selectivity of conjugates of the present invention.
- Fig. 8 is a schematic representation of the cleavage of the peptide linkage of Fig. 5 within a target cell endosome.
- Fig. 9 shows anti-toxin reactivity of normal human serum samples. Normal human donor sera were tested for reactivity with diphtheria toxin (DT) , Pseudomonas exotoxin A (PE) , pertussis holotoxin (PTx) and pertussis B oligomer (PTxB) .
- Fig. 10 schematically depicts antigen processing of a protein antigen (i.e.. intact toxin) to small peptides that associate with MHC Class II antigens.
- Fig. 11 shows the inhibition of proliferation in a mixed lymphocyte reaction (MLR) by cholera holotoxin (CTx) and cholera B oligomer (CTxB) (measured by H-thymidine incorporation) .
- MLR mixed lymphocyte reaction
- CTx cholera holotoxin
- CTxB cholera B oligomer
- Fig. 12 shows a controlled charge modification reaction scheme which protects lysine groups of toxins that are important in biological function, while reducing the immunogenicity of conjugates of the present invention.
- BRM Bio Response Modifier
- Cytokine and Lymphokine A moiety, e.g.. a protein, that is capable of eliciting a functional response from target cells measurable in an in vitro or in vivo bioassay.
- a biological response might include alterations in differentiation functions, activation, induction or suppression of proliferation, inhibition or enhancement of secretion or de novo synthesis or alterations in synthesis of proteins, and the like.
- a BRM may enhance or suppress one or more of these functional responses.
- Target Cells A defined population of cells from which a biological response is to be elicited.
- Retargeted Toxin A toxin wherein the endogenous cell binding portion has been replaced by a different targeting moiety exhibiting preferential binding to target cells.
- the "replacement" is accomplished by biochemical or recombinant means and includes deleting, deactivating, blocking or otherwise nullifying the native BRM cell binding portion and conjugating the resultant toxin moiety by biochemical or recombinant means to a new targeting agent.
- Non-Cytolytic toxins For the purposes of this specification, the term "non-cytolytic toxin" describes a protein toxin whose predominant affect is not to directly inhibit protein synthesis.
- Non-cytolytic toxins affect non-essential or differentiation functions carried out by target cells. Since non-cytolytic toxins do not affect protein synthesis, such moieties do not directly cause cell death in most circumstances.
- An exemplary exception to this generalization is the process of apocytosis. The death of some T-cells, for example, is metabolically pre-programmed, and the program can be initiated by increased cAMP levels. Consequently, it might be expected that the generally non-cytolytic toxins that affect cAMP levels in target cells may cause the death of T-cells when targeted thereto.
- non-cytolytic toxins or conjugates thereof may halt proliferation of normally proliferating cells, such as tumor cells. Some of these non- proliferating cells may then die as a result of their inability to proliferate. Reduction in tumor size may result from inhibition of proliferation since tumor cell growth is a product of two processes, cell proliferation and death.
- Tables V and VI A listing of exemplary non- cytolytic toxins, their substrates, and mechanism of action appears in Tables V and VI.
- Cytolytic Toxin A protein toxin moiety that inhibits protein synthesis, and is therefore directly capable of killing targeted as well as non-targeted cells demonstrating non-specific uptake. Conjugates derived from these toxins will demonstrate dose- limiting toxicities as a consequence of antigen- specific or non-specific uptake. Exemplary sources of such toxin moieties include plants (ricin, abrin, pokeweed antiviral protein, gelonin, and modecin) , bacteria (diphtheria, Pseudomonas exotoxin A, and shigella) , and marine organisms.
- a protein, peptide, or non- proteinaceous organic molecule that preferentially binds to a population of cells intended for pharmaceutical manipulation i.e.. target cells
- a targeting moiety may, for example, be a growth factor (e.g.. and epidermal growth factor, a transforming growth factor, such as alpha or beta) , a hormone, a cytocline, an antibody, a colony stimulating factor, transferrin, bombesin, gastrin releasing peptide or the like.
- the targeting moiety may bind to a protein, glycoprotein, or glycolipid located on the target cell surface or an internal antigen.
- target cells are antibodies, fragments or functional equivalents thereof, such as those binding to the NR-LU-10 (an IgG 2b murine monoclonal antibody directed to the 37-40 kilodalton pancarcinoma glycoprotein) , NR-ML- 05 (IgG 2b ,k i munoglobulin may be generated in accordance with Woodhouse et al.. "In Human Melanoma, From Basic Research to Clinical Application, "
- Ferrine ed., Springer-Verlag, pp. 151-163, 1990
- NR-CE-01 antigens ⁇ .CEA
- growth factors such as EGF, TGF ⁇ , TGF ⁇ , FGF, transferrin, interleukins 1-11, GM-CSF and G-CSF.
- Preferred targeting agents useful in this regard are proteins, including antibody and antibody fragments; peptides, such as bombesin, gastrin- releasing peptide, cell adhesion peptides, substance P, neuromedin-B, neuromedin-C, and metenkephalin; and hormones, such as EGF, c.-, and ⁇ -TGF, estradiol, neurotensin, melanocyte stimulating hormone, follicle stimulating hormone, luteinizing hormone, and human growth hormone.
- proteins including antibody and antibody fragments
- peptides such as bombesin, gastrin- releasing peptide, cell adhesion peptides, substance P, neuromedin-B, neuromedin-C, and metenkephalin
- hormones such as EGF, c.-, and ⁇ -TGF, estradiol, neurotensin, melanocyte stimulating hormone, follicle stimulating hormone, luteinizing hormone, and human growth hormone.
- Biotin, avidin, proteins corresponding to known cell surface receptors (including low density lipoproteins, transferrin and insulin) , fibrinolytic enzymes, and biological response modifiers (including interleukin, interferon, erythropoietin and colony-stimulating factor) are also preferred targeting agents.
- Analogs of the above-listed targeting agents that retain the capacity to bind to a defined target cell population may also be used within the claimed invention.
- synthetic proteinaceous targeting agents may be designed.
- a first BRM for example, is functionally equivalent to a second BRM if the BRM function of the first is achieved by the second. Consequently, non-cytolytic toxin conjugates capable of inducing a functional response from a target cell population are functionally equivalent, regardless of origin.
- a non-cytolytic toxin BRM purified from a natural source and conjugated to a targeting moiety is functionally equivalent to such a BRM conjugate formed from a fusion protein.
- a holotoxin having an inactivated or occluded cell binding domain retargeted by a targeting moiety is functionally equivalent to a non- cytolytic toxin portion devoid of endogenous cell binding specificity.
- a holotoxin, domain- sized or multiple domain-sized toxin portions, or a small peptide derived from or corresponding to a toxin portion of less than domain size that is purified from a native toxin or prepared by recombinant techniques may also be functionally equivalent when used in conjugates of the present invention.
- Substrate The moiety within target cells acted on by the non-cytolytic enzymatic toxin portion of the present invention.
- ATP Adenosine tri-phosphate.
- ADP Adenosine di-phosphate.
- cAMP Cyclic adenosine mono-phosphate.
- GDP Guanosine di-phosphate.
- GTP Guanosine tri-phosphate.
- Such toxin portions may be of bacterial, marine, or plant origin or may be manufactured by recombinant techniques.
- Bacterial toxin moieties of the present invention are or may be derived from, for example, enterotoxins or exotoxins and may be of one or a multiple chain structure.
- a complete or whole toxin with both enzymatic and cell binding activity is referred to as a holotoxin.
- Toxin moieties need only exhibit a non-cytolytic function that results in alteration of a BRM activity or differentiation or slowing of cellular proliferation when appropriately targeted to a population of target cells to be useful in the present invention.
- Cytolytic, protein synthesis-inhibiting toxins such as ricin, diphtheria, or Pseudomonas exotoxin A exhibit commonalities in function, but that functional commonality is not necessarily reflected by identical mechanisms of action or structural or sequence similarity.
- Such toxins include an enzymatic or catalytic portion, a translocating portion, and a cell binding portion.
- the enzymatic portion constitutes the toxicity generating portion of the toxin, i.e.. catalyzes a modification of regulatory proteins or processes within a cell leading to inhibition of protein synthesis and eventually to cell death.
- cytolytic toxin is responsible for the internalization of that toxin into the target cell and transfer of the enzymatic subunit to the cytoplasm.
- the cell binding portion of the cytolytic toxin dictates the endogenous cell binding specificity of that toxin. The latter two functions may be present within the same polypeptide or subunit.
- non-cytolytic toxins express these three functional activities associated with protein domains.
- the enzymatic domain or subunit must be included in conjugates of the present invention to impart BRM activity thereto.
- an appropriate enzymatic domain may, for example, either amplify or block the signal by interacting with G-proteins, thereby either enhancing or halting the biological response triggered by the ligand.
- Similar enzymatically active portions can be derived from non-cytolytic toxins that impact the metabolic processes of target cells directly without interacting through G-proteins. Such directly acting toxin moieties (e.g..).
- ADP ribosylation reactions are central to the metabolic signaling mechanisms manipulated by retargeted conjugates of some aspects of the present invention.
- ADP ribosylation of certain substrates involved in the operation of intracellular signaling mechanisms results in the amplification or inhibition of target cell functional processes.
- Exemplary substrates for ADP-ribosylation by bacterial or plant toxins are G-proteins, elongation factor-2 (EF-2) , actin, rho-like oncogene proteins, and the like. Only toxins that act on EF-2 have been previously employed as portions of conjugates, however.
- toxin-substrate relationships that may be exploited by the present invention are shown in Tables V and VI are discussed below.
- Cholera toxin and E. coli heat labile enterotoxin cause ADP ribosylation of the G-protein alpha s.
- the endogenous cell binding specificity of cholera toxin is to GM-1 ganglioside, a receptor found on a plethora of cell types.
- E. coli heat labile enterotoxin exhibits endogenous cell binding specificity for GM-1, GDlb, and asialo-GM-1.
- Pertussis toxin induces ADP ribosylation of G-protein alpha i and exhibits endogenous cell binding specificity for glycoproteins that are ubiquitous on mammalian cells.
- Botulinum neurotoxins act on ras-like proto- oncogene substrates. Ras oncogenes are similar to G- alpha proteins in function and may regulate tumor cell proliferation. A current theory endeavoring to explain this similarity is that such oncogenes are mutated G-proteins. Specifically, ras oncogenes are thought to be irreversibly activated G-proteins. Consequently, inhibition of ras function may lead to growth arrest of tumor cells.
- Botulinum neurotoxins exhibit endogenous cell binding specificity for many gangliosides. Conjugates or fusion proteins of these toxins affecting G-proteins or modified G-proteins and targeting moieties constitute an aspect of the present invention.
- this aspect of the present invention is directed to conjugates including a non- cytolytic toxin moiety and a ligand coupled to an effector through G-proteins and capable of triggering a desired biological response.
- Non-cytolytic toxin moieties useful in conjugates of the first aspect of the present invention are toxin moieties that can alter a cellular function by modification of G-proteins and thus the existing target cell signaling mechanism.
- Exemplary non-cytolytic toxin moieties are portions of toxins, such as cholera toxin Al subunit and pertussis toxin SI subunit, in which the enzymatic function of the respective holotoxins reside.
- non-cytolytic toxin moieties act on G-proteins, and thus on cellular signaling mechanisms which utilize receptors and effectors coupled through G-proteins. Functional processes subject to G-protein control therefore can be regulated by a non-cytolytic toxin portion of cholera or pertussis toxin.
- the non-cytolytic portion of cholera toxin induces ADP-ribosylation of a G-alpha protein designated the stimulatory alpha protein (G-alpha s) .
- Active G-alpha s GTP form
- GTP form may then interact with adenyl cyclase, inducing an increase in cAMP level.
- the activation signal is down regulated by enzymatic degradation of GTP to GDP, restoring the G-protein subunit to its inactive GDP form.
- Extended activation or suppression of G-protein mediated response may be caused by ADP ribosylation of the G-protein subunit by the non-cytolytic toxin.
- Increases in cAMP level may impact many target cell metabolic processes, such as inhibiting proliferation.
- G-alpha protein designated either G-alpha i or G-alpha o.
- effector moiety function is no longer resultant from the ligand-receptor interaction. Under these conditions, ligand-receptor binding does not result in enhanced adenyl cyclase activity. Consequently, intracellular cAMP levels are not increased.
- exemplary biochemical and recombinant technique-generated cholera toxin conjugates of the present invention are shown schematically in Figs. 3 and 4.
- the structure of cholera toxin is shown schematically in Fig. 3.
- the toxin subunit exhibiting enzymatic activity is designated "Al, " and is linked through a disulfide bond to a smaller peptide "A2.”
- Al enzymatic toxin portion
- TP targeting moiety
- Non-cytolytic toxins that do not act on G- proteins are also useful in the present invention.
- Such toxins are capable of directly modulating cyclic nucleotide levels (second aspect of the present invention) or modifying cellular actin (third aspect of the present invention) .
- Conjugates or fusion proteins of the second aspect of the present invention involve, for example, adenylate cyclase toxins of Bordetella pertussis and Bacillus anthraces that have enzymatic activity functionally similar to mammalian adenyl cyclase. Insertion of this toxin protein within target cells will thus act directly on ATP to increase intra ⁇ cellular cAMP levels. Such toxins can thereby affect functional activities regulated by cAMP. The toxicity of such bacterial enzymes is directed to peripheral leukocytes, namely PMNs, but retargeting will reduce this otherwise dose-limiting toxicity.
- adenylate cyclase toxin of Bordetella pertussis exhibit catalyzing, translocating, and cell binding functions.
- the catalytic (i.e. , enzymatic) function requires mammalian calmodulin for activity and is associated with a domain located between amino acids 1 and 450.
- the translocation function is likely provided by a toxin domain located between amino acids 450 and 1000.
- the cell binding function is likely provided by a toxin domain located between amino acids 1000 and 1600.
- the toxin has protease-sensitive sites in the structural region between amino acid 450 and amino acid 1600.
- adenylate cyclase toxin of Bordetella pertussis are schematically shown in Fig. 6.
- the longer derivative corresponds to the enzymatic and translocating domains of the adenylate cyclase toxin, while the shorter derivative includes only the enzymatic domain.
- the impact of target cell protease activity on the conjugates is reduced by use of the shorter derivative, thereby prolonging the effective duration of conjugate activity.
- the potency of a conjugate containing a toxin portion devoid of the translocating domain may be less than that of a holotoxin.
- each described moiety will vary according to the targeting molecule; for example, a rapidly internalizing targeting molecule complex will be more active than one that internalizes slowly when using the molecule devoid of the translocating domain.
- the smaller derivative can be utilized with an artificial translocating peptide specified by Copending U.S. Patent Application No. 07/232,337, entitled “Covalently-Linked Complexes and Methods for Enhanced Cytotoxicity and Imaging, filed August 15, 1988. Conjugation of the adenyl cyclase toxin portion and targeting moiety is preferably achieved through the peptide linkers described herein, or as fusion proteins.
- protease sensitive site(s) in native or recombinant Bordetella pertussis toxin or portions thereof could be identified and modified by site-specific mutagenesis, so that those sites would no longer be cleaved by target cell proteases.
- the protease-sensitive sites may be deleted.
- B. anthraces toxin appears to internalize within cells through a receptor-mediated event.
- the B. anthraces toxin may be more useful for conjugation, as the enzymatic subunit may be more easily separated from cell binding activity.
- Actin polymerization is important to a number of cellular processes including secretion (i.e.. exocytosis) and proliferation.
- ADP ribosylation of actin has been shown to inhibit actin polymerization.
- Botulinum C2 toxins and Clostridial iota toxins act on (i.e.. induce ADP ribosylation of actin) , thereby more directly affecting target cell function (i.e.. effect achieved with no G-protein or modified G-protein intermediary) .
- the endogenous cell binding specificity of these toxins is unknown.
- the third aspect of the present invention features conjugates or fusion proteins of non-cytolytic toxins capable of ADP ribosylation of actin and targeting moieties.
- conjugates of the second and third aspects need not be targeted with ligands capable of triggering responses mediated through G-proteins.
- ligands capable of triggering responses mediated through G-proteins.
- a broader array of ligands or targeting proteins can be utilized to target the cell population in which the conjugates will effect a biological response upon internaliza- tion.
- the biological response(s) elicited will be a function of the target cells and the differentiation functions that those cells carry out.
- the inclusion of the translocating domain of a non-cytolytic toxin in conjugates of the present invention may be necessary to allow full conjugate potency.
- the rate and degree of target cell internalization and egress from target cell endosomes into the cytoplasm could be enhanced by the toxin translocating domain.
- holotoxins i.e.. whole toxins
- Conjugation of holotoxins to targeting moieties must be achieved in a manner such that the endogenous cell binding portion of the holotoxin is at least partially occluded in some manner, thereby preventing the endogenous toxin cell binding portion from interacting with endogenous target cells.
- a conjugate of the present invention is the ability to release the toxin portion from endosomal vesicles into the target cell cytoplasm, allowing it to interact with its substrate.
- a releasing mechanism may be already present in the structure of a holotoxin.
- the sequence active in release is found near the enzymatic subunit or domain.
- diphtheria toxin likely contains such a sequence, since a portion roughly corresponding to the enzymatic domain is released into the cytoplasm while the B chain is required for optimal conjugate potency (binding and translocation) .
- Similar findings have been made with respect to Pseudomonas exotoxin A and cholera toxin. In the latter case, a proteolytically sensitive state may exist in the A2 peptide adjacent to the enzymatic Al subunit.
- deletion variants of the B-chain in combination with the enzymatic domain are constructed and tested for potency.
- fusion proteins or chemical conjugates of such deletion variants may be constructed and similarly tested. In this ' manner, the smallest portion of the B-chain able to confer optimal potency is identified. Since optimal potency requires the release mechanism, one also simultaneously identifies the portion(s) of the B chain with the sensitive sequence.
- a linker is preferably provided to separate the targeting moiety and toxin portion and allow the targeting moiety to bind to its receptor.
- Conjugates of the present invention may there- fore be linked by disulfide and thioether linkages will as well as by peptide bonds.
- An exemplary linking procedure involves two steps: (1) reacting the toxin portion and the targeting moiety with the same or different hetero or homobifunctional cross- linker to form a derivatized toxin portion and a derivatized targeting moiety, and (2) reacting the derivatized toxin portion and the derivatized targeting moiety to obtain a covalently-linked conjugate.
- Conjugate species are then purified from excess chemical linker or unreacted toxin and targeting moiety by one or more of a variety of chromatographic procedures. A practitioner in the art could design and implement appropriate conjugation and separation procedures.
- native sulfhydryl groups located on the toxin portion or the targeting moiety may be reduced, for example, with DTT (dithiothreitol) to form derivatized moieties for use in step (2) described above.
- a sulfhydryl moiety may be introduced into the targeting moiety and a maleimide moiety may be introduced into the toxin portion through derivatization of lysine groups with a heterobiofunctional reagent. A condensation reaction of such derivatized components may then be undertaken to form conjugates of the present invention. See, for example, U.S. Patent No. 4,981,979.
- the following commercially available agents may be used to derivatize the toxin BRMs and/or targeting moieties of conjugates of the present invention: Succinimidyl 4- (N-maleimidomethyl) cyclohexane- 1-carboxylate (SMCC) .
- SMCC Succinimidyl 4- (N-maleimidomethyl) cyclohexane- 1-carboxylate
- Sulfosuccinimidyl 4- (N-maleimidomethyl) cyclohexane-1-carboxylate (Sulfo-SMCC) .
- MBS m-maleimidobenzoyl-N-hydroxysuccinimide ester
- Sulfo-MBS m-maleimidobenzoyl-N-hydroxysulfosuccinimide ester
- N-succinimidyl 3- (2-pyridyldithio) propionate SPDP
- succinimidyl 4- p-maleimidophenyl butyrate (SMPD)
- N-succinimidyl (4-iodoacetyl) aminobenzoate (SIAB) .
- Sulfosuccinimidyl (4-iodoacetyl) aminobenzoate Sulfo-SIAB
- Recombinant procedures may be employed to provide fusion proteins of non-cytolytic toxin moieties and a proteinaceous ligand or targeting agent.
- Exemplary fusion protein production methods are illustrated in Bailon et al., Biotechnology, 6:1326, 1988; Chaudhary et al. , Proc. Nat'l Acad. Sci. (USA) 84:4538, 1987; Siena et al., Blood. 72:756, 1988; Chaudhary et al. , Proc. Nat'l Acad. Sci. (USA) . 87:1066, 1990; Chaudhary et al. , Nature. 339:394, 1989; Pastan and Fitzgerald, J. Biol.
- Disulfide linkages may be prematurely cleaved in vivo prior to delivery of the conjugate to target cells, however.
- a preferred peptide linker useful for constructing fusion proteins in accordance with the present invention is shown in Fig. 7.
- a toxin enzymatic portion encoded by a nucleotide sequence within a fusion protein and expressed in an appropriate prokaryotic or eukaryotic system and designated "A, " is bound to a targeting moiety designated "T.P.” by a sequence encoding a peptide linker.
- the peptide linker includes residues capable of specifying flanking spacer regions, terminal glycine residues, and a disulfide bond loop containing a consensus sequence sensitive to endosomal or lysomal enzymes.
- the peptide linker both separates the enzymatic subunit from the targeting moiety and provides for enzymatic subunit release upon target cell internalization.
- Amino acid residues sensitive to lysosomal and endosomal protease cleavage are known.
- An example of such a sequence is given by the single amino acid code KWVEE.
- the indicated sequence can be cleaved by a variety of endosomal or lysosomal enzymes including cathepsins A through H.
- the availability of the sequence to enzymes located within target cell endosomes is enhanced by the presence of that sequence within a disulfide bonded loop.
- the disulfide bond is protected from premature reduction by liver disulfide isomerase or by serum glutathione by the peptide sequence which is insensitive to such serum enzymes.
- the entire cholera toxin A2 sequence may be encoded into the fusion protein instead of the simpler peptide sequence. Based on previous data, this is the site likely to be the endogenous site of cleavage within cholera toxin.
- the release of the biological response modifying toxin portion A within target cell endosomes is shown schematically in Fig. 8.
- a conjugate or fusion protein of the present invention is internalized within a target cell endosome, the conjugate is subjected to low pH, a reducing environment and endosomal proteases such as cathepsins. Endosome proteases cleave the sensitive peptide linker. The disulfide bond portion of the dual linker then undergoes reduction allowing release of the enzymatic domain of the conjugates of the present invention from the targeting moiety.
- non-cytolytic toxins such as those capable of interacting with target cell metabolic signaling mechanisms, exhibit cell binding specificity for a variety of target cells.
- the portion of the toxin responsible for cell binding is removed and substituted with a targeting moiety having a receptor that is enriched on the target cell population.
- the increase in bioavailability may alternatively be achieved by inactivation or blockage of the endogenous cell binding specificity and toxin retargeting with a more application-appropriate targeting moiety.
- the retargeted conjugates of the first aspect of the present invention only induce biological responses within receptor positive cells. In addition, these conjugates will only affect differentiation rather than essential cellular functions within target cells.
- the conjugates of the first aspect of the present invention are non-toxic to target cells.
- Non-receptor bearing cells that are non-specifically targeted by conjugates of the present invention would not likely be affected at all, because a receptor-triggered event must occur in combination with an enzymatic modification of G-proteins to produce the biological response. Internalization by pino- or phagocytosis of conjugate would not induce a response, as signaling would not have been triggered by ligand binding.
- cholera toxin binds to GM-1 ganglioside, a moiety present on many normal tissues.
- the toxicity of cholera toxin is, however, only manifested after binding to GM-1 ganglioside located on colonic epithelium. Such binding induces active water resorption and acute diarrhea.
- Removal or inactivation of the cholera toxin B chain eliminates endogenous binding and the adverse effects thereof.
- Substituting IL-2 for the B chain allows targeting of cholera toxin to IL-2 receptor positive T-cells. Since IL-2 receptors are not located on the colonic epithelial cells, no dose- limiting diarrhea will be exhibited by the conjugate.
- a bacterial toxin such as Pseudomonas exotoxin A (PE)
- PE Pseudomonas exotoxin A
- Fig. 9 shows that, although most individuals have not been exposed or vaccinated to PE, occasional "normal" humans have pre-existing anti-PE antibodies. For both pertussis toxin and PE, pre-existing antibody is therefore generally of little concern. In contrast, pre-existing antibody would be of concern if one utilized diphtheria toxin in conjugates of the present invention.
- mice models may be used to estimate the immunogenicity threshold for a toxin conjugate. For example, about 2 ⁇ g of pertussis toxin B oligomer plus adjuvant has been shown to be necessary to invoke an optimal immune response in a mouse model. Lower doses that are sufficient to induce a biological response will therefore be poorly immunogenic. Chronic administration over a period of months, however, will likely induce an immune response.
- portions of a toxin molecule are deleted through recombinant methods so as to eliminate non-BRM active portions of the molecule.
- the third approach features identification and modification of the specific epitope(s) of the toxin molecule predominantly responsible for the anti-toxin response. For instance, immunodominant sites of
- Pseudomonas exotoxin A and pertussis toxin have been associated with the enzymatic subunit or domain. Genetic manipulation may be used to alter (by deletion or site-specific mutagenesis, for example) these immunodominant sites within the enzymatic subunit, resulting in a toxin derivative possessing reduced immunogenicity. The resulting mutagenized toxin would still be immunogenic, but would likely not induce as rapid or as high-titered responses as native toxin using the same dose of immunogen.
- the fourth approach contemplates administration of immunogenic unmodified toxin to a "modified recipient" population that is unable to generate a normal and vigorous immune response (for example, patients that are immunosuppressed due to disease such as AIDS, and/or treatment such as cancer chemotherapy) .
- the fifth approach involves chemical treatment that modifies certain amino acid residues of the toxin, thereby producing a derivative that elicits little or no anti-toxin response while retaining functional BRM activity.
- Exemplary methods in this regard are described in co-pending U.S. Patent Application Serial Nos. 07/157,213 and 07/330,846, hereby incorporated by reference in their entireties.
- An additional approach involves derivatizing the toxin or fusion protein with polyethyleneglycol. Cationization of proteins has an enhancing effect on antigen presentation and immunogenicity.
- anionic modification of proteins may be employed to create a relatively homogeneous distribution of negative charges on the surface of the toxin. Such anionic modification may reduce the ability of macrophages to metabolize and present degradative peptides to immunocompetent cells.
- toxins having a homogeneous charge distribution may not be well-recognized by T-cell dependent mechanisms. These consequences of charge modification result in a relative decrease in toxin immunogenicity.
- Toxins undergo antigen processing when injected into a mammalian recipient. Antigen processing involves degradation of the intact toxin molecule to small peptides (Fig. 10) . These small peptides associate with MHC Class II (HLA-DR) antigens of the recipient's immune system on the surface of recipient's antigen presenting cells. The small peptides must "fit" the HLA-DR structure (i.e.. have the proper amino acid sequence to interact with appropriate amino acids within the HLA-DR antigen) .
- HLA-DR MHC Class II
- immunodominance may vary with the haplotype of the individual recipient, especially in outbred populations (i.e.. humans) . Consequently, identifi- cation of consensus immunodominant sequences may be difficult. Modification of specific amino acids or sequences to reduce immunogenicity may therefore be so complex as to become infeasible.
- An exemplary means for reducing toxin immuno ⁇ genicity through chemical modification involves treatment of the toxin with succinic anhydride.
- Succinic anhydride can modify the following amino acid residues under appropriate conditions: lysine, histidine, tryptophan, serine, threonine, and cysteine (free SH form) .
- Amino acid residues believed to be essential for biological/chemical activities of ADP-ribosylating toxins include lysine (cell binding and possibly translocation) , glutamic acid (NAD binding) , tyrosine (NAD binding) and histidine or tryptophan (substrate binding) .
- a histidine residue essential for substrate binding is inaccessible in the absence of toxin unfolding, so succinic anhydride exposure would not modify this essential residue.
- Glutamic acid is also not modified by succinic anhydride.
- the lysine residues of PE are more problematic. A practitioner in the art, however, would be able to minimize lysine modification through alteration of pH and reactant levels.
- lysine protecting and deprotecting steps can be used in a succinylating methodology, such as that discussed in Example II below, to overcome this problem.
- efficacious anti-tumor agents have been developed which act to inhibit cAMP regulated cellular processes such as proliferation, differentiation, or the process of metastasis.
- Conjugates of two aspects of the present invention are useful for modulating the intracellular target cell cAMP level and provide for high potency and relatively long serum half life compared to small molecules.
- target cells are tumor cells
- elevated cAMP levels will result in inhibition of proliferation or induction of differentiation.
- anti-tumor effects are known to be elicited by agents that modulate cAMP levels in a stoichiometric fashion, including the alkylating inhibitor 8-C1- cAMP, agonists or antagonists of phosphoinositol production, chemical inhibitors of cAMP dependent protein kinase (PKA) and antisense oligonucleotides for inhibition of PKA.
- PKA cAMP dependent protein kinase
- these stoichiometric agents are relatively non-toxic. Since tumor cells are abnormal, different therapeutic approaches than those envisioned for normal cells should be contemplated for treatment including conjugates of the second aspect of the present invention. Tumor cells often undergo de- differentiation upon neoplastic conversion. Normal signal transduction and/or receptors may be lost in this process. Exemplary of such a loss upon neoplastic conversion is the p21 H-ras oncogene. This oncogene bears a resemblance to a G-alpha protein; however, p21 H-ras oncogene exists permanently in the active state.
- the tumor cells could be targeted with a conjugate utilizing targeting moieties such as epidermal growth factor (EGF) , transforming growth factor, such as alpha or beta, transferrin bombesin, gastrin releasing peptide or monoclonal antibodies specific to tumor cells and an enzymatically active toxin moiety derived from the adenyl cyclase toxins of B. pertussis or B. anthraces.
- GEF epidermal growth factor
- transforming growth factor such as alpha or beta
- transferrin bombesin gastrin releasing peptide or monoclonal antibodies specific to tumor cells
- gastrin releasing peptide or monoclonal antibodies specific to tumor cells
- an enzymatically active toxin moiety derived from the adenyl cyclase toxins of B. pertussis or B. anthraces.
- Increasing the level of intracellular cAMP can be achieved in this manner, thereby causing differentiation or halting proliferation
- Conjugates of the second aspect of the present invention achieve an elevation in intracellular cAMP level independently of G-protein mediated signaling processes and, thus, are not subject to alterations in signal transduction mechanisms resulting from neoplastic transformation.
- Certain advantages are garnered by the use of conjugates capable of elevating cyclic nucleotide levels rather than conventional stoichiometric (i.e.. chemical) regulators thereof.
- conjugates capable of elevating cyclic nucleotide levels rather than conventional stoichiometric (i.e.. chemical) regulators thereof.
- such conjugates have a higher potency than agents which act stoichiometricly since they are enzymatic or catalytic in nature.
- Conjugates of the second aspect of the present invention are expected to induce a target cell metabolic response at a concentration of about 10 " M (in vitro) , based on the demonstrated potency of conjugates that utilize cytolytic toxins.
- stoichiometric modulators of cAMP levels such as forskolin
- conjugates of the present invention, especially of the first aspect generate a metabolic effect of prolonged duration (e.g.. hours to days as opposed to minutes) .
- stoichiometric modulators usually express activity for only as long as sufficient levels are maintained at the cellular or tissue level.
- conjugates of the present invention may be formed of a relevant toxin domain and a chemical or proteinaceous targeting moiety specific for a certain population of target cells.
- the conjugates of the present invention may be designed to induce a specific metabolic response in a specific target cell population, thereby allowing flexibility in therapeutic design and known mechanism of action.
- Stoichiometric modulators on the other hand, have required extensive research to determine structure-function relationships and mechanisms of action and are relatively non-specific.
- tumor cell proliferation can result in cell death.
- an increase in tumor cAMP level has been shown to inhibit the ability of the cells to metastasize.
- Increased cAMP levels have also been shown to increase chemotherapeutic drug accumulation in tumor cells through the reversal of drug resistance.
- Patients with germ cell malignancies have been treated with cytotoxic chemotherapy (generally, cis-platinum, vinblastine and bleomycin) . Although the intent of this treatment is to kill the cells, in some patients neoplastic tumors differentiate into benign tumors that are then excised. These patients are often cured.
- non-cytolytic, retargeted toxins of the present invention can induce differentiation in the tumor cells. Tumor cells are poorly immunogenic.
- Tumor cells that differentiate may induce or express higher levels of differentiation antigens.
- differentiation antigens can be highly immunogenic.
- the cancer patient's humoral and cellular immune system could mount a greater response to the differentiated tumor cells than to the undifferentiated cells.
- Differentiation of tumor cells can also be a strategy to enhance the efficiency of other therapies.
- the estrogen receptor status of breast carcinoma has been shown to be an independent prognostic variable. More importantly, the level of expression of this receptor on tumor cells predicts the response of the tumor to treatment with tamoxifen, a synthetic anti-estrogen. Breast tumors with high estrogen receptor expression respond much more frequently to tamoxifen treatment than those with low or absent levels of the receptor.
- a retargeted toxin of the present invention may be utilized to cause differentiation of the tumor cells rendering them more estrogen-dependent, permitting the tumor cells to respond adequately to subsequent tamoxifen treatment.
- Another example is the HER-2/neu oncoprotein whose expression is increased in poor prognosis breast cancer.
- An antibody/non-cytolytic toxin conjugate or fusion protein of the present invention can be used to target these cells and cause them to differentiate or retard their proliferation.
- Associated Antigens may also be upregulated as a result of a cAMP level increase in tumor target cells. Upregulation of TAA could improve anti-TAA antibody binding to the tumor cell. Consequently, the effectiveness of passive administration of diagnostic or therapeutic antibodies will be enhanced. For example, therapeutic or diagnostic conjugates employing antibody targeting agents will bind at higher levels to differentiated tumor cells, thereby enhancing the therapeutic or diagnostic effect. Upregulation of receptors can also enhance receptor-ligand binding. Subsequent therapies involving radioactive or otherwise conjugated ligand may be rendered more effective in this manner.
- conjugates of the first aspect of the present invention designed to increase the intracellular level of cAMP of tumor cells through modification of proteins and signaling mechanisms, may be efficacious against tumor cells that have maintained a normal signaling mechanism.
- the existence of such a normally functioning, metabolic signaling mechanism must, however, be established and shown to be maintained within multiple tumors of the same histological type and capable of eliciting differentiation or proliferation inhibition.
- conjugates or fusion proteins of cholera toxin Al (enzymatic subunit) and EGF, TGF- alpha, transferrin, or antibody to TAA may be employed to induce differentiation or inhibition of proliferation of tumor target cells.
- Such conjugates need to be shown to induce differentiation or inhibition of proliferation in vitro.
- Effective conjugates are subsequently evaluated in animal models of neoplastic disease using a therapeutic protocol (i.e.. taxomifen therapy of breast cancer) to determine in vivo efficacy.
- a therapeutic protocol i.e.. taxomifen therapy of breast cancer
- toxin BRM conjugates of the present invention having proliferative activity may be advantageously used, singly or in combination with other agents, for the treatment of AIDS or HIV infection.
- Pertussis toxin has been known to induce a lymphocytosis.
- the enzymatic activity of the toxin is required for this purpose.
- Recent data indicates that while pertussis toxin is capable of stimulating the proliferation and subsequent maturation of both CD-4 and CD-8 positive T-cells, the latter predominate in long term culture.
- an IL-7 targeted pertussis SI enzymatic subunit conjugate or fusion protein may be utilized. Retargeting eliminates the normal dose-limiting toxicity and provides a BRM known to act on early lymphoid progenitors.
- Enzymatic modification of G-proteins in such progenitors enhances the proliferative activity of IL-7 resulting in increased production in the bone marrow and, with maturation in the thymus, increased levels of CD-4 positive T-cells in the periphery.
- conjugates or fusion proteins are administered in conjunction with an agent capable of inhibiting virus replication, such as azathiopyrimidme (AZT) , BI-RG-587, CD-4 peptides or functional equivalents thereof, the combination therapy will lead to a recovery of virus negative CD-4 positive T-cells and elevated CD-4/CD-8 ratios.
- combination therapy involving a conjugate or fusion protein of the present invention and an HIV infection inhibiting agent, such as CD-4/Ig6, might be employed.
- CD5-targeted pertussis or cholera toxin conjugate Another potential therapeutic application involves a CD5-targeted pertussis or cholera toxin conjugate.
- CD-5 though expressed on virtually all T-cells, is present also on a subpopulation of B- cells, which are continuously renewing and likely responsible for production of certain auto-immune antibodies and perhaps auto-immune diseases. Inhibiting proliferation of such cells or engendering a decrease in auto-immune immunoglobulin may thus lead to the resolving of auto-immune disease.
- early success has been reported in treating rheumatoid arthritis with a CD-5/ricin A chain conjugate.
- This conjugate as is typical of conjugates utilizing portions of cytolytic toxins, demonstrates dose limiting hepatic toxicity.
- conjugates of CD-5 and pertussis SI or cholera Al could inhibit proliferation of CD-5 positive B-cells without giving rise to hepatic toxicity.
- the conjugates of the present invention are intended for injection into humans or other mammalian recipients. Accordingly, appropriate manufacturing and in vitro storage practices must be observed to provide suitable sterile, pyrogen-free compositions. Although not necessary, it is preferable to use a pharmaceutically acceptable extender or filler to dilute any carrier which might optionally be used and/or to simplify metering the requisite small quantities of such compounds.
- Sodium chloride and glucose are preferred carriers; sodium chloride is especially preferred because it facilitates provision of an isotonic solution.
- An effective dose of a conjugate of the present invention is an amount sufficient to elicit a therapeutically meaningful biological response in a recipient.
- An appropriate dose would range from about 1 milligram to about 10 grams and be administered by intravenous, intraperitoneal, subcutaneous, intramuscular, or other routes of administration.
- the exact dosage and administration route will vary depending on the nature of the patient's ailment, conjugate components used, and the like. A practitioner in the medical arts would be able to determine an appropriate dose and administration route for conjugates of the present invention.
- the following Examples are presented for illustrative purposes only and do not therefore limit the scope of the present invention in any way.
- Cholera toxin has been previously described to be suppressive for lectin-stimulated proliferation of peripheral blood mononuclear cells in vitro. In addition, recent studies have indicated that cholera toxin can enhance renal allograft survival.
- Fig. 11 shows the performance of cholera holotoxin (CTx) and the B oligomer of cholera toxin (CTxB) with respect to the inhibition of MLR proliferation.
- CTx cholera holotoxin
- CTxB B oligomer of cholera toxin
- Cholera toxin has been shown to be both able to abrogate proliferation of T-cells and bind to colonic epithelium, causing acute diarrhea. Consequently, cholera toxin cannot be administered at a dose sufficient to achieve optimal immunosuppression of T- cells. Removal of the B-chain and retargeting the toxin A-chain with IL-2, i.e.. interleukin-2, would allow specific suppression of activated T-cells without dose-limiting diarrhea.
- IL-2 i.e. interleukin-2
- CTxAl Cholera toxin Al subunit
- the mixture is passed over a PD-10 gel filtration column (Pharmacia) equilibrated in 0.15M NaCl, 0.1M sodium phosphate (pH 7.5) (PBS) to remove unreacted SPDP.
- IL-2 is similarly reacted with SPDP.
- Derivatized CTxAl is then treated with 50 mM dithiothreitol (DTT) to generate a free SH moiety and then desalted.
- Derivatized and reduced CTxAl is mixed with derivatized IL-2 and allowed to react at room temperature for a time period extending from about 6 hours to overnight.
- conjugate is then chromatographed on a TSK-3000 column (Waters) using an FPLC system (Pharmacia) to separate conjugate from unreacted materials. Conjugate-containing fractions are then pooled and may then be used, for example, for in vivo allograft rejection studies or treatment.
- the CTxAl starting material or a conjugate or fusion protein thereof produced in Example I may be subjected to a controlled chemical modification reaction scheme as shown in Fig. 13.
- the proteinaceous starting material is incubated with citraconic anhydride under conditions sufficient to selectively modify easily accessible lysine residues (i.e.. 10-50:1 offering citraconic anhydride to protein ratio, pH 9-9.5).
- the modified protein is then reacted with succinic anhydride at neutral pH and at high offering (excess of succinic anhydride) to modify all other susceptible residues.
- the protein is then dialyzed in phosphate buffer at pH 5.0, allowing hydrolysis of the lysine adducts and restoration of the original epsilon amino functionality. Modification control and extensive succinylation are achieved using this immunogenicity reducing method, while the BRM activity of the conjugate, dependent on lysines, is retained.
- Pseudomonas exotoxin A is very immunogenic in both mice and humans, and both types of recipients respond to the same immunodominant site(s) of the toxin molecule (Morgan, unpublished information) .
- Morgan unpublished information
- To examine the immunogenicity of unmodified vs. succinylated PE BALB/c mice were injected with 1 ⁇ g/mouse NR-LU-10/PE conjugate (unmodified) , NR-LU-10-PE conjugate (toxin was succinylated prior to conjugation) or HSA.
- Anti-PE titers were determined by solid phase ELISA at days 0, 14 and 28.
- NR-LU-10 is an antibody that recognizes a 37-40 kd pan-carcinoma glycoprotein. Mice injected with NR-LU-10-PE conjugate demonstrated a high titer anti-PE response at day 14 post-injection (Table IX) . Control mice (HSA administered) showed no anti-PE response. TABLE IX
- succinylation of PE significantly reduces the immunogenicity thereof in mice.
- immunogenicity of other bacterial toxins may be reduced by succinylation.
- a conjugate of epidermal growth factor (EGF) and an adenylate cyclase toxin portion derived from B. anthraces is prepared in accordance with previously described recombinant procedures.
- EGF epidermal growth factor
- a patient bearing a tumor which is EGF positive will receive a single, daily pharmacologically effective dose of the conjugate by an intravenous administration route.
- the conjugate is delivered to tumor cells, the adenylate cyclase toxin is internalized therein.
- This BRM is expected to catalyze the conversion of ATP to cAMP.
- proliferation of the tumor cells Upon the increase in intracellular cAMP levels, proliferation of the tumor cells will be inhibited and/or the cells will be caused to differentiate, resulting in tumor stasis.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Engineering & Computer Science (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Chemical & Material Sciences (AREA)
- Molecular Biology (AREA)
- Immunology (AREA)
- Cell Biology (AREA)
- Medicinal Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Epidemiology (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Peptides Or Proteins (AREA)
Abstract
Conjugués utiles pour modifier des fonctions de cellules cibles afin d'obtenir des résultats thérapeutiques. Les conjugués décrits peuvent comprendre une toxine non cytolytique qui n'inhibe pas directement la synthèse de protéine et qui peut agir par l'intermédiaire d'un mécanisme existant de signalisation de métabolisme cellulaire; ainsi qu'une fraction de ciblage qui constitue un ligand reconnu par le récepteur de cellules cibles présent dans le mécanisme de signalisation existant. Dans une variante, les conjugués de la présente invention peuvent comprendre un domaine de toxine pouvant influencer directement un processus métabolique des cellule cibles (par exemple, catalyser la conversion d'ATP en cAMP) ou agissant sur un substrat intervenant dans ces processus (par exemple, l'actine); ainsi qu'une fraction de ciblage présentant une spécificité pour la population de cellules cibles. On décrit des procédés d'utilisation de ces conjugués.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US74515891A | 1991-08-15 | 1991-08-15 | |
US745,158 | 1991-08-15 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO1993004191A1 true WO1993004191A1 (fr) | 1993-03-04 |
Family
ID=24995499
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US1992/006823 WO1993004191A1 (fr) | 1991-08-15 | 1992-08-13 | Conjugues de toxines non cytolytiques |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO1993004191A1 (fr) |
Cited By (13)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
EP0752885A4 (fr) * | 1992-09-25 | 1996-02-06 | Neorx Corp | Inhibiteur therapeutique de cellules des muscles vasculaires lisses |
WO1998046255A1 (fr) * | 1997-04-17 | 1998-10-22 | Yale University | Inhibition d'etats immunitaires pathologiques a mediation par les lymphocytes b-1 |
US6395513B1 (en) | 1995-04-21 | 2002-05-28 | The Speywood Laboratory, Ltd. | Clostridial toxin derivatives able to modify peripheral sensory afferent functions |
EP1350523A2 (fr) * | 1992-09-25 | 2003-10-08 | Neorx Corporation | Conjugués thérapeutiques inhibitant les cellules des muscles lisses vasculaires |
US6632440B1 (en) | 1998-08-25 | 2003-10-14 | Health Protection Agency | Methods and compounds for the treatment of mucus hypersecretion |
US7052702B1 (en) | 1997-10-08 | 2006-05-30 | Health Protection Agency | Conjugates of galactose-binding lectins and clostridial neurotoxins as analgesics |
US7192596B2 (en) | 1996-08-23 | 2007-03-20 | The Health Protection Agency Ipsen Limited | Recombinant toxin fragments |
US7208466B1 (en) | 1999-03-31 | 2007-04-24 | The Health Protection Agency | Use of a lectin or conjugates for modulation of c-fibre activity |
US7625410B2 (en) | 2001-05-02 | 2009-12-01 | Boston Scientific Scimed, Inc. | Stent device and method |
US7674470B2 (en) | 1996-08-23 | 2010-03-09 | Health Protection Agency | Recombinant toxin fragments |
US7727538B2 (en) | 1998-08-25 | 2010-06-01 | Syntaxin Ltd. | Methods and compounds for the treatment of mucus hypersecretion |
US8012491B2 (en) | 1996-08-23 | 2011-09-06 | Syntaxin, Ltd. | Recombinant toxin fragments |
US8852603B2 (en) | 1999-09-23 | 2014-10-07 | Syntaxin Limited | Inhibition of secretion from non-neuronal cells |
Citations (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US4664911A (en) * | 1983-06-21 | 1987-05-12 | Board Of Regents, University Of Texas System | Immunotoxin conjugates employing toxin B chain moieties |
-
1992
- 1992-08-13 WO PCT/US1992/006823 patent/WO1993004191A1/fr active Application Filing
Patent Citations (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US4664911A (en) * | 1983-06-21 | 1987-05-12 | Board Of Regents, University Of Texas System | Immunotoxin conjugates employing toxin B chain moieties |
Non-Patent Citations (2)
Title |
---|
J. EXP. MED., Volume 167, issued February 1988, BACHA et al., "Interleukin 2 Receptor-Targeted Cytotoxicity: Interleukin 2 Receptor-Mediated Action of a Diphtheria Toxin-Related Interleukin 2 Fusion Protein", pages 612-622. * |
PROC. NATL. ACAD. SCI. USA, Volume 83, issued November 1986, MURPHY et al., "Genetic Construction, Expression, and Melanoma-Selective Cytotoxicity of a Diphtheria Toxin-Related Alpha-Melanocyte-Stimulating Hormone Fusion Protein". * |
Cited By (25)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
EP0752885A1 (fr) * | 1992-09-25 | 1997-01-15 | Neorx Corporation | Inhibiteur therapeutique de cellules des muscles vasculaires lisses |
EP1350523A2 (fr) * | 1992-09-25 | 2003-10-08 | Neorx Corporation | Conjugués thérapeutiques inhibitant les cellules des muscles lisses vasculaires |
EP0752885A4 (fr) * | 1992-09-25 | 1996-02-06 | Neorx Corp | Inhibiteur therapeutique de cellules des muscles vasculaires lisses |
EP1350523A3 (fr) * | 1992-09-25 | 2003-12-10 | Neorx Corporation | Conjugués thérapeutiques inhibitant les cellules des muscles lisses vasculaires |
US7887810B2 (en) | 1995-04-21 | 2011-02-15 | Syntaxin, Ltd | Clostridial toxin derivatives able to modify peripheral sensory afferent functions |
US8158132B2 (en) | 1995-04-21 | 2012-04-17 | Syntaxin Limited | Clostridial toxin derivatives able to modify peripheral sensory afferent functions |
US6395513B1 (en) | 1995-04-21 | 2002-05-28 | The Speywood Laboratory, Ltd. | Clostridial toxin derivatives able to modify peripheral sensory afferent functions |
US6962703B2 (en) | 1995-04-21 | 2005-11-08 | Ipsen Limited | Clostridial toxin derivatives able to modify peripheral sensory afferent functions |
US7892560B2 (en) | 1995-04-21 | 2011-02-22 | Syntaxin, Ltd | Clostridial toxin derivatives able to modify peripheral sensory afferent functions |
US8012491B2 (en) | 1996-08-23 | 2011-09-06 | Syntaxin, Ltd. | Recombinant toxin fragments |
US8012479B2 (en) | 1996-08-23 | 2011-09-06 | Health Protection Agency | Recombinant toxin fragments |
US8454976B2 (en) | 1996-08-23 | 2013-06-04 | Syntaxin Limited | Recombinant toxin fragments |
US8017134B2 (en) | 1996-08-23 | 2011-09-13 | Syntaxin Limited | Recombinant toxin fragments |
US7674470B2 (en) | 1996-08-23 | 2010-03-09 | Health Protection Agency | Recombinant toxin fragments |
US7897158B2 (en) | 1996-08-23 | 2011-03-01 | Syntaxin, Ltd | Recombinant toxin fragments |
US7192596B2 (en) | 1996-08-23 | 2007-03-20 | The Health Protection Agency Ipsen Limited | Recombinant toxin fragments |
WO1998046255A1 (fr) * | 1997-04-17 | 1998-10-22 | Yale University | Inhibition d'etats immunitaires pathologiques a mediation par les lymphocytes b-1 |
US7052702B1 (en) | 1997-10-08 | 2006-05-30 | Health Protection Agency | Conjugates of galactose-binding lectins and clostridial neurotoxins as analgesics |
US7452543B2 (en) | 1997-10-08 | 2008-11-18 | Syntaxin Ltd. | Conjugates of galactose-binding lectins and clostridial neurotoxins as analgesics |
US7727538B2 (en) | 1998-08-25 | 2010-06-01 | Syntaxin Ltd. | Methods and compounds for the treatment of mucus hypersecretion |
US6632440B1 (en) | 1998-08-25 | 2003-10-14 | Health Protection Agency | Methods and compounds for the treatment of mucus hypersecretion |
US7208466B1 (en) | 1999-03-31 | 2007-04-24 | The Health Protection Agency | Use of a lectin or conjugates for modulation of c-fibre activity |
US8852603B2 (en) | 1999-09-23 | 2014-10-07 | Syntaxin Limited | Inhibition of secretion from non-neuronal cells |
US7625410B2 (en) | 2001-05-02 | 2009-12-01 | Boston Scientific Scimed, Inc. | Stent device and method |
US9006395B2 (en) | 2002-09-12 | 2015-04-14 | The Secretary Of State For Health | Recombinant toxin fragments |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
AU2001275246B2 (en) | T cell receptor fusions and conjugates and methods of use thereof | |
Smith et al. | Glycosphingolipids as toxin receptors | |
US20200172594A1 (en) | T cell receptor fusions and conjugates and methods of use thereof | |
US8252897B2 (en) | Modified toxins | |
AU720857B2 (en) | Use of toxin peptides and/or affinity handles for delivery compounds into cells | |
US6962694B1 (en) | Cytolysis of target cells by superantigen conjugates inducing T-cell activation | |
US11136367B2 (en) | Multi-level specific targeting of cancer cells | |
WO1993004191A1 (fr) | Conjugues de toxines non cytolytiques | |
AU2001275246A1 (en) | T cell receptor fusions and conjugates and methods of use thereof | |
NO333676B1 (no) | Nytt konstruert superantigen for human terapi | |
JP4296536B2 (ja) | Gb3レセプター発現細胞を標的とする分子の一般担体 | |
US20230405129A1 (en) | Proinflammatory prodrugs | |
US5571507A (en) | Methods of treating diabetes | |
AU632995B2 (en) | Cell specific cytotoxic agents | |
WO1993015751A1 (fr) | TOXINES CHIMERIQUES SE FIXANT AU RECEPTEUR DE GnRH | |
US5681810A (en) | Diphtheria toxin fragments, conjugates and methods of use to inhibit tumors and leukemia | |
AU697469B2 (en) | Intracellular delivery of chemical agents to a specific cell type | |
CA2389302A1 (fr) | Distribution de proteines a travers des couches de cellules epitheliales polaires | |
Brinkmann | Recombinant immunotoxins: protein engineering for cancer therapy | |
CA2247432A1 (fr) | Promedicaments macromoleculaire de ciblage des lymphocytes t | |
US5621078A (en) | Modified pseudomonas exotoxin PE40 | |
Gilliland et al. | Chimeric toxins containing fragment A from diphtheria toxin | |
Foss | Fusion Toxins | |
JPH0219324A (ja) | 卵巣細胞に対するモノクローナル抗体 |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
AK | Designated states |
Kind code of ref document: A1 Designated state(s): CA JP |
|
AL | Designated countries for regional patents |
Kind code of ref document: A1 Designated state(s): AT BE CH DE DK ES FR GB GR IE IT LU MC NL SE |
|
122 | Ep: pct application non-entry in european phase | ||
NENP | Non-entry into the national phase |
Ref country code: CA |