WO2022177677A1 - Egfr de domaine iv tronqué et utilisations correspondantes - Google Patents
Egfr de domaine iv tronqué et utilisations correspondantes Download PDFInfo
- Publication number
- WO2022177677A1 WO2022177677A1 PCT/US2022/012621 US2022012621W WO2022177677A1 WO 2022177677 A1 WO2022177677 A1 WO 2022177677A1 US 2022012621 W US2022012621 W US 2022012621W WO 2022177677 A1 WO2022177677 A1 WO 2022177677A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- egfr
- seq
- antibody
- sequence
- set forth
- Prior art date
Links
- 108060006698 EGF receptor Proteins 0.000 title description 54
- 210000004027 cell Anatomy 0.000 claims abstract description 432
- 210000001744 T-lymphocyte Anatomy 0.000 claims abstract description 60
- 238000001727 in vivo Methods 0.000 claims abstract description 20
- 230000002163 immunogen Effects 0.000 claims abstract description 14
- 102000052116 epidermal growth factor receptor activity proteins Human genes 0.000 claims abstract 18
- 108700015053 epidermal growth factor receptor activity proteins Proteins 0.000 claims abstract 18
- YOHYSYJDKVYCJI-UHFFFAOYSA-N n-[3-[[6-[3-(trifluoromethyl)anilino]pyrimidin-4-yl]amino]phenyl]cyclopropanecarboxamide Chemical compound FC(F)(F)C1=CC=CC(NC=2N=CN=C(NC=3C=C(NC(=O)C4CC4)C=CC=3)C=2)=C1 YOHYSYJDKVYCJI-UHFFFAOYSA-N 0.000 claims abstract 18
- 150000007523 nucleic acids Chemical class 0.000 claims description 130
- 102000039446 nucleic acids Human genes 0.000 claims description 111
- 108020004707 nucleic acids Proteins 0.000 claims description 111
- 238000009739 binding Methods 0.000 claims description 107
- 230000027455 binding Effects 0.000 claims description 106
- 206010028980 Neoplasm Diseases 0.000 claims description 93
- 238000000034 method Methods 0.000 claims description 87
- 150000001413 amino acids Chemical group 0.000 claims description 63
- 201000011510 cancer Diseases 0.000 claims description 61
- 239000000427 antigen Substances 0.000 claims description 46
- 125000001151 peptidyl group Chemical group 0.000 claims description 37
- 239000013604 expression vector Substances 0.000 claims description 35
- 108010019670 Chimeric Antigen Receptors Proteins 0.000 claims description 34
- 239000000203 mixture Substances 0.000 claims description 32
- 239000013598 vector Substances 0.000 claims description 27
- 230000011664 signaling Effects 0.000 claims description 26
- 239000011230 binding agent Substances 0.000 claims description 18
- 102000003812 Interleukin-15 Human genes 0.000 claims description 11
- 108090000172 Interleukin-15 Proteins 0.000 claims description 11
- 230000003834 intracellular effect Effects 0.000 claims description 11
- 230000001177 retroviral effect Effects 0.000 claims description 11
- 102000003675 cytokine receptors Human genes 0.000 claims description 7
- 108010057085 cytokine receptors Proteins 0.000 claims description 7
- 102000004022 Protein-Tyrosine Kinases Human genes 0.000 claims description 6
- 108090000412 Protein-Tyrosine Kinases Proteins 0.000 claims description 6
- 108091008874 T cell receptors Proteins 0.000 claims description 6
- 102000016266 T-Cell Antigen Receptors Human genes 0.000 claims description 6
- 238000000338 in vitro Methods 0.000 claims description 6
- 210000001778 pluripotent stem cell Anatomy 0.000 claims description 2
- 108090000623 proteins and genes Proteins 0.000 abstract description 144
- 239000003550 marker Substances 0.000 abstract description 10
- 230000001404 mediated effect Effects 0.000 abstract description 10
- 230000001225 therapeutic effect Effects 0.000 abstract description 10
- 230000001413 cellular effect Effects 0.000 abstract description 7
- 108010047041 Complementarity Determining Regions Proteins 0.000 description 373
- 239000012634 fragment Substances 0.000 description 253
- 102000004169 proteins and genes Human genes 0.000 description 115
- 235000018102 proteins Nutrition 0.000 description 113
- 230000010056 antibody-dependent cellular cytotoxicity Effects 0.000 description 77
- 235000001014 amino acid Nutrition 0.000 description 63
- 229940024606 amino acid Drugs 0.000 description 62
- 230000000694 effects Effects 0.000 description 60
- 125000003275 alpha amino acid group Chemical group 0.000 description 58
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 57
- 102000001301 EGF receptor Human genes 0.000 description 55
- 229960005395 cetuximab Drugs 0.000 description 54
- 125000003729 nucleotide group Chemical group 0.000 description 54
- 239000002773 nucleotide Substances 0.000 description 52
- 108090000765 processed proteins & peptides Proteins 0.000 description 47
- 201000010099 disease Diseases 0.000 description 45
- 102100035360 Cerebellar degeneration-related antigen 1 Human genes 0.000 description 44
- 108091007433 antigens Proteins 0.000 description 43
- 102000036639 antigens Human genes 0.000 description 43
- 239000012636 effector Substances 0.000 description 39
- 101100112922 Candida albicans CDR3 gene Proteins 0.000 description 38
- 102100035361 Cerebellar degeneration-related protein 2 Human genes 0.000 description 38
- 101000737796 Homo sapiens Cerebellar degeneration-related protein 2 Proteins 0.000 description 38
- 101000737793 Homo sapiens Cerebellar degeneration-related antigen 1 Proteins 0.000 description 36
- 102000004196 processed proteins & peptides Human genes 0.000 description 35
- 238000006467 substitution reaction Methods 0.000 description 34
- 239000003446 ligand Substances 0.000 description 33
- 229920001184 polypeptide Polymers 0.000 description 33
- 241000699670 Mus sp. Species 0.000 description 28
- 210000003819 peripheral blood mononuclear cell Anatomy 0.000 description 27
- 102000005962 receptors Human genes 0.000 description 27
- 108020003175 receptors Proteins 0.000 description 27
- 241000699666 Mus <mouse, genus> Species 0.000 description 24
- 239000000126 substance Substances 0.000 description 24
- 229940116977 epidermal growth factor Drugs 0.000 description 23
- 238000011282 treatment Methods 0.000 description 23
- 102000009465 Growth Factor Receptors Human genes 0.000 description 22
- 108010009202 Growth Factor Receptors Proteins 0.000 description 22
- 102100033237 Pro-epidermal growth factor Human genes 0.000 description 22
- 230000000295 complement effect Effects 0.000 description 20
- 238000000684 flow cytometry Methods 0.000 description 20
- 239000012528 membrane Substances 0.000 description 20
- 108020004414 DNA Proteins 0.000 description 19
- 238000002474 experimental method Methods 0.000 description 19
- 239000000047 product Substances 0.000 description 19
- 230000008685 targeting Effects 0.000 description 19
- 238000003556 assay Methods 0.000 description 16
- 108091028043 Nucleic acid sequence Proteins 0.000 description 15
- 230000006870 function Effects 0.000 description 15
- 108091033319 polynucleotide Proteins 0.000 description 15
- 102000040430 polynucleotide Human genes 0.000 description 15
- 239000002157 polynucleotide Substances 0.000 description 15
- 238000010361 transduction Methods 0.000 description 15
- 230000026683 transduction Effects 0.000 description 15
- -1 e.g. Chemical class 0.000 description 14
- 230000001603 reducing effect Effects 0.000 description 14
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 13
- 230000001745 anti-biotin effect Effects 0.000 description 13
- 150000001875 compounds Chemical class 0.000 description 13
- 238000011534 incubation Methods 0.000 description 13
- 238000002360 preparation method Methods 0.000 description 13
- 239000000523 sample Substances 0.000 description 13
- 208000024891 symptom Diseases 0.000 description 13
- 238000001890 transfection Methods 0.000 description 13
- 210000004881 tumor cell Anatomy 0.000 description 13
- 238000002965 ELISA Methods 0.000 description 12
- 101150029707 ERBB2 gene Proteins 0.000 description 12
- 229940098773 bovine serum albumin Drugs 0.000 description 12
- 208000035475 disorder Diseases 0.000 description 12
- 239000003814 drug Substances 0.000 description 12
- 239000000499 gel Substances 0.000 description 12
- 238000012360 testing method Methods 0.000 description 12
- 102100024222 B-lymphocyte antigen CD19 Human genes 0.000 description 11
- 238000004422 calculation algorithm Methods 0.000 description 11
- 210000004408 hybridoma Anatomy 0.000 description 11
- 230000005764 inhibitory process Effects 0.000 description 11
- 230000003612 virological effect Effects 0.000 description 11
- 101000980825 Homo sapiens B-lymphocyte antigen CD19 Proteins 0.000 description 10
- 241001465754 Metazoa Species 0.000 description 10
- 230000004913 activation Effects 0.000 description 10
- 238000007792 addition Methods 0.000 description 10
- 125000000539 amino acid group Chemical group 0.000 description 10
- 239000003795 chemical substances by application Substances 0.000 description 10
- 108020001507 fusion proteins Proteins 0.000 description 10
- 102000037865 fusion proteins Human genes 0.000 description 10
- 238000004020 luminiscence type Methods 0.000 description 10
- 230000009467 reduction Effects 0.000 description 10
- 102220003351 rs387906411 Human genes 0.000 description 10
- 230000000903 blocking effect Effects 0.000 description 9
- 230000003247 decreasing effect Effects 0.000 description 9
- 238000001943 fluorescence-activated cell sorting Methods 0.000 description 9
- 230000002401 inhibitory effect Effects 0.000 description 9
- 238000004519 manufacturing process Methods 0.000 description 9
- 239000000463 material Substances 0.000 description 9
- 239000002105 nanoparticle Substances 0.000 description 9
- 210000000822 natural killer cell Anatomy 0.000 description 9
- 229920000642 polymer Polymers 0.000 description 9
- 238000000746 purification Methods 0.000 description 9
- 230000032258 transport Effects 0.000 description 9
- 229960000575 trastuzumab Drugs 0.000 description 9
- 238000005406 washing Methods 0.000 description 9
- HBZBAMXERPYTFS-SECBINFHSA-N (4S)-2-(6,7-dihydro-5H-pyrrolo[3,2-f][1,3]benzothiazol-2-yl)-4,5-dihydro-1,3-thiazole-4-carboxylic acid Chemical compound OC(=O)[C@H]1CSC(=N1)c1nc2cc3CCNc3cc2s1 HBZBAMXERPYTFS-SECBINFHSA-N 0.000 description 8
- 102000017420 CD3 protein, epsilon/gamma/delta subunit Human genes 0.000 description 8
- 108091026890 Coding region Proteins 0.000 description 8
- 108020004705 Codon Proteins 0.000 description 8
- WHUUTDBJXJRKMK-UHFFFAOYSA-N Glutamic acid Natural products OC(=O)C(N)CCC(O)=O WHUUTDBJXJRKMK-UHFFFAOYSA-N 0.000 description 8
- 101000851181 Homo sapiens Epidermal growth factor receptor Proteins 0.000 description 8
- 101000914514 Homo sapiens T-cell-specific surface glycoprotein CD28 Proteins 0.000 description 8
- 230000006044 T cell activation Effects 0.000 description 8
- 102100027213 T-cell-specific surface glycoprotein CD28 Human genes 0.000 description 8
- 230000000692 anti-sense effect Effects 0.000 description 8
- 230000001086 cytosolic effect Effects 0.000 description 8
- 238000012217 deletion Methods 0.000 description 8
- 230000037430 deletion Effects 0.000 description 8
- 235000013922 glutamic acid Nutrition 0.000 description 8
- 239000004220 glutamic acid Substances 0.000 description 8
- 239000003112 inhibitor Substances 0.000 description 8
- 239000011325 microbead Substances 0.000 description 8
- 238000012986 modification Methods 0.000 description 8
- 239000013612 plasmid Substances 0.000 description 8
- 239000000243 solution Substances 0.000 description 8
- 238000002198 surface plasmon resonance spectroscopy Methods 0.000 description 8
- 206010006187 Breast cancer Diseases 0.000 description 7
- 208000026310 Breast neoplasm Diseases 0.000 description 7
- 101000917858 Homo sapiens Low affinity immunoglobulin gamma Fc region receptor III-A Proteins 0.000 description 7
- 108060001084 Luciferase Proteins 0.000 description 7
- 239000005089 Luciferase Substances 0.000 description 7
- 108091005461 Nucleic proteins Proteins 0.000 description 7
- 239000002552 dosage form Substances 0.000 description 7
- 238000005259 measurement Methods 0.000 description 7
- 230000004048 modification Effects 0.000 description 7
- 230000001105 regulatory effect Effects 0.000 description 7
- 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 6
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 6
- 101000917839 Homo sapiens Low affinity immunoglobulin gamma Fc region receptor III-B Proteins 0.000 description 6
- 108060003951 Immunoglobulin Proteins 0.000 description 6
- 102100029193 Low affinity immunoglobulin gamma Fc region receptor III-A Human genes 0.000 description 6
- 206010033128 Ovarian cancer Diseases 0.000 description 6
- 108090000873 Receptor Protein-Tyrosine Kinases Proteins 0.000 description 6
- IQFYYKKMVGJFEH-XLPZGREQSA-N Thymidine Chemical compound O=C1NC(=O)C(C)=CN1[C@@H]1O[C@H](CO)[C@@H](O)C1 IQFYYKKMVGJFEH-XLPZGREQSA-N 0.000 description 6
- 238000004458 analytical method Methods 0.000 description 6
- 210000004899 c-terminal region Anatomy 0.000 description 6
- 210000000170 cell membrane Anatomy 0.000 description 6
- 239000003153 chemical reaction reagent Substances 0.000 description 6
- 230000007423 decrease Effects 0.000 description 6
- 239000003623 enhancer Substances 0.000 description 6
- 239000001963 growth medium Substances 0.000 description 6
- 102000018358 immunoglobulin Human genes 0.000 description 6
- 238000009169 immunotherapy Methods 0.000 description 6
- 230000003993 interaction Effects 0.000 description 6
- 239000002502 liposome Substances 0.000 description 6
- 239000002609 medium Substances 0.000 description 6
- 238000011275 oncology therapy Methods 0.000 description 6
- 230000003405 preventing effect Effects 0.000 description 6
- 230000008569 process Effects 0.000 description 6
- 241000894007 species Species 0.000 description 6
- 238000010186 staining Methods 0.000 description 6
- 241000283707 Capra Species 0.000 description 5
- 241000124008 Mammalia Species 0.000 description 5
- 206010061535 Ovarian neoplasm Diseases 0.000 description 5
- 108010076504 Protein Sorting Signals Proteins 0.000 description 5
- 102000004278 Receptor Protein-Tyrosine Kinases Human genes 0.000 description 5
- 102000004142 Trypsin Human genes 0.000 description 5
- 108090000631 Trypsin Proteins 0.000 description 5
- 239000005557 antagonist Substances 0.000 description 5
- 239000002246 antineoplastic agent Substances 0.000 description 5
- 239000011324 bead Substances 0.000 description 5
- 239000000872 buffer Substances 0.000 description 5
- 238000002512 chemotherapy Methods 0.000 description 5
- 239000012228 culture supernatant Substances 0.000 description 5
- 230000001186 cumulative effect Effects 0.000 description 5
- 229940079593 drug Drugs 0.000 description 5
- 238000005516 engineering process Methods 0.000 description 5
- 238000009472 formulation Methods 0.000 description 5
- 239000010931 gold Substances 0.000 description 5
- 102000045108 human EGFR Human genes 0.000 description 5
- 210000002865 immune cell Anatomy 0.000 description 5
- 210000004263 induced pluripotent stem cell Anatomy 0.000 description 5
- 208000032839 leukemia Diseases 0.000 description 5
- 239000004005 microsphere Substances 0.000 description 5
- 230000035772 mutation Effects 0.000 description 5
- 239000008194 pharmaceutical composition Substances 0.000 description 5
- 238000001542 size-exclusion chromatography Methods 0.000 description 5
- 210000001519 tissue Anatomy 0.000 description 5
- 239000012588 trypsin Substances 0.000 description 5
- 239000003981 vehicle Substances 0.000 description 5
- 239000013603 viral vector Substances 0.000 description 5
- 238000001262 western blot Methods 0.000 description 5
- 102000053602 DNA Human genes 0.000 description 4
- 229910052688 Gadolinium Inorganic materials 0.000 description 4
- DHMQDGOQFOQNFH-UHFFFAOYSA-N Glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 description 4
- NYHBQMYGNKIUIF-UUOKFMHZSA-N Guanosine Chemical compound C1=NC=2C(=O)NC(N)=NC=2N1[C@@H]1O[C@H](CO)[C@@H](O)[C@H]1O NYHBQMYGNKIUIF-UUOKFMHZSA-N 0.000 description 4
- 241000282412 Homo Species 0.000 description 4
- ZDXPYRJPNDTMRX-VKHMYHEASA-N L-glutamine Chemical compound OC(=O)[C@@H](N)CCC(N)=O ZDXPYRJPNDTMRX-VKHMYHEASA-N 0.000 description 4
- FFEARJCKVFRZRR-BYPYZUCNSA-N L-methionine Chemical group CSCC[C@H](N)C(O)=O FFEARJCKVFRZRR-BYPYZUCNSA-N 0.000 description 4
- 206010025323 Lymphomas Diseases 0.000 description 4
- 238000011579 SCID mouse model Methods 0.000 description 4
- PXIPVTKHYLBLMZ-UHFFFAOYSA-N Sodium azide Chemical compound [Na+].[N-]=[N+]=[N-] PXIPVTKHYLBLMZ-UHFFFAOYSA-N 0.000 description 4
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 4
- KZSNJWFQEVHDMF-UHFFFAOYSA-N Valine Natural products CC(C)C(N)C(O)=O KZSNJWFQEVHDMF-UHFFFAOYSA-N 0.000 description 4
- 230000001594 aberrant effect Effects 0.000 description 4
- 230000009471 action Effects 0.000 description 4
- 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 4
- 238000013459 approach Methods 0.000 description 4
- 239000012472 biological sample Substances 0.000 description 4
- 229960002685 biotin Drugs 0.000 description 4
- 239000011616 biotin Substances 0.000 description 4
- 239000002775 capsule Substances 0.000 description 4
- 239000000969 carrier Substances 0.000 description 4
- 230000003833 cell viability Effects 0.000 description 4
- 239000010949 copper Substances 0.000 description 4
- 230000008878 coupling Effects 0.000 description 4
- 238000010168 coupling process Methods 0.000 description 4
- 238000005859 coupling reaction Methods 0.000 description 4
- OPTASPLRGRRNAP-UHFFFAOYSA-N cytosine Chemical compound NC=1C=CNC(=O)N=1 OPTASPLRGRRNAP-UHFFFAOYSA-N 0.000 description 4
- 210000001151 cytotoxic T lymphocyte Anatomy 0.000 description 4
- 238000011161 development Methods 0.000 description 4
- 230000004069 differentiation Effects 0.000 description 4
- 239000002158 endotoxin Substances 0.000 description 4
- 230000004927 fusion Effects 0.000 description 4
- UIWYJDYFSGRHKR-UHFFFAOYSA-N gadolinium atom Chemical compound [Gd] UIWYJDYFSGRHKR-UHFFFAOYSA-N 0.000 description 4
- 230000001900 immune effect Effects 0.000 description 4
- 238000003018 immunoassay Methods 0.000 description 4
- 230000001965 increasing effect Effects 0.000 description 4
- 238000003780 insertion Methods 0.000 description 4
- 230000037431 insertion Effects 0.000 description 4
- 238000007912 intraperitoneal administration Methods 0.000 description 4
- 150000002500 ions Chemical class 0.000 description 4
- 230000002147 killing effect Effects 0.000 description 4
- 230000000670 limiting effect Effects 0.000 description 4
- 229910052751 metal Inorganic materials 0.000 description 4
- 239000002184 metal Substances 0.000 description 4
- 229930182817 methionine Chemical group 0.000 description 4
- 239000002777 nucleoside Substances 0.000 description 4
- ZJAOAACCNHFJAH-UHFFFAOYSA-N phosphonoformic acid Chemical class OC(=O)P(O)(O)=O ZJAOAACCNHFJAH-UHFFFAOYSA-N 0.000 description 4
- 230000002829 reductive effect Effects 0.000 description 4
- 239000012146 running buffer Substances 0.000 description 4
- 150000003839 salts Chemical class 0.000 description 4
- 239000003826 tablet Substances 0.000 description 4
- RWQNBRDOKXIBIV-UHFFFAOYSA-N thymine Chemical compound CC1=CNC(=O)NC1=O RWQNBRDOKXIBIV-UHFFFAOYSA-N 0.000 description 4
- 238000013518 transcription Methods 0.000 description 4
- 230000035897 transcription Effects 0.000 description 4
- 238000012546 transfer Methods 0.000 description 4
- 239000004474 valine Substances 0.000 description 4
- NHBKXEKEPDILRR-UHFFFAOYSA-N 2,3-bis(butanoylsulfanyl)propyl butanoate Chemical compound CCCC(=O)OCC(SC(=O)CCC)CSC(=O)CCC NHBKXEKEPDILRR-UHFFFAOYSA-N 0.000 description 3
- JKMHFZQWWAIEOD-UHFFFAOYSA-N 2-[4-(2-hydroxyethyl)piperazin-1-yl]ethanesulfonic acid Chemical compound OCC[NH+]1CCN(CCS([O-])(=O)=O)CC1 JKMHFZQWWAIEOD-UHFFFAOYSA-N 0.000 description 3
- 241000251468 Actinopterygii Species 0.000 description 3
- DWRXFEITVBNRMK-UHFFFAOYSA-N Beta-D-1-Arabinofuranosylthymine Natural products O=C1NC(=O)C(C)=CN1C1C(O)C(O)C(CO)O1 DWRXFEITVBNRMK-UHFFFAOYSA-N 0.000 description 3
- 241000283690 Bos taurus Species 0.000 description 3
- VYZAMTAEIAYCRO-UHFFFAOYSA-N Chromium Chemical compound [Cr] VYZAMTAEIAYCRO-UHFFFAOYSA-N 0.000 description 3
- 102000004190 Enzymes Human genes 0.000 description 3
- 108090000790 Enzymes Proteins 0.000 description 3
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 3
- 108010021625 Immunoglobulin Fragments Proteins 0.000 description 3
- 102000008394 Immunoglobulin Fragments Human genes 0.000 description 3
- 102100025390 Integrin beta-2 Human genes 0.000 description 3
- 108091092195 Intron Proteins 0.000 description 3
- 238000010824 Kaplan-Meier survival analysis Methods 0.000 description 3
- QNAYBMKLOCPYGJ-REOHCLBHSA-N L-alanine Chemical compound C[C@H](N)C(O)=O QNAYBMKLOCPYGJ-REOHCLBHSA-N 0.000 description 3
- 229930182816 L-glutamine Natural products 0.000 description 3
- COLNVLDHVKWLRT-QMMMGPOBSA-N L-phenylalanine Chemical compound OC(=O)[C@@H](N)CC1=CC=CC=C1 COLNVLDHVKWLRT-QMMMGPOBSA-N 0.000 description 3
- KZSNJWFQEVHDMF-BYPYZUCNSA-N L-valine Chemical compound CC(C)[C@H](N)C(O)=O KZSNJWFQEVHDMF-BYPYZUCNSA-N 0.000 description 3
- 108091034117 Oligonucleotide Proteins 0.000 description 3
- 102000035195 Peptidases Human genes 0.000 description 3
- 108091005804 Peptidases Proteins 0.000 description 3
- 206010035226 Plasma cell myeloma Diseases 0.000 description 3
- 108010008281 Recombinant Fusion Proteins Proteins 0.000 description 3
- 102000007056 Recombinant Fusion Proteins Human genes 0.000 description 3
- 241000283984 Rodentia Species 0.000 description 3
- 108091081021 Sense strand Proteins 0.000 description 3
- 108010003723 Single-Domain Antibodies Proteins 0.000 description 3
- 235000004279 alanine Nutrition 0.000 description 3
- 230000004075 alteration Effects 0.000 description 3
- 230000000259 anti-tumor effect Effects 0.000 description 3
- 230000009286 beneficial effect Effects 0.000 description 3
- IQFYYKKMVGJFEH-UHFFFAOYSA-N beta-L-thymidine Natural products O=C1NC(=O)C(C)=CN1C1OC(CO)C(O)C1 IQFYYKKMVGJFEH-UHFFFAOYSA-N 0.000 description 3
- 230000015572 biosynthetic process Effects 0.000 description 3
- 235000020958 biotin Nutrition 0.000 description 3
- 230000022534 cell killing Effects 0.000 description 3
- 230000005754 cellular signaling Effects 0.000 description 3
- 230000008859 change Effects 0.000 description 3
- 238000012512 characterization method Methods 0.000 description 3
- 239000013522 chelant Substances 0.000 description 3
- 238000006243 chemical reaction Methods 0.000 description 3
- 229910052804 chromium Inorganic materials 0.000 description 3
- 239000011651 chromium Substances 0.000 description 3
- 239000002299 complementary DNA Substances 0.000 description 3
- 230000000139 costimulatory effect Effects 0.000 description 3
- 230000001472 cytotoxic effect Effects 0.000 description 3
- 238000001514 detection method Methods 0.000 description 3
- 238000010586 diagram Methods 0.000 description 3
- 230000002222 downregulating effect Effects 0.000 description 3
- 230000002708 enhancing effect Effects 0.000 description 3
- 230000002255 enzymatic effect Effects 0.000 description 3
- 229940088598 enzyme Drugs 0.000 description 3
- 125000000291 glutamic acid group Chemical group N[C@@H](CCC(O)=O)C(=O)* 0.000 description 3
- PCHJSUWPFVWCPO-UHFFFAOYSA-N gold Chemical compound [Au] PCHJSUWPFVWCPO-UHFFFAOYSA-N 0.000 description 3
- 229910052737 gold Inorganic materials 0.000 description 3
- 230000028993 immune response Effects 0.000 description 3
- 210000000987 immune system Anatomy 0.000 description 3
- 230000001024 immunotherapeutic effect Effects 0.000 description 3
- 230000000415 inactivating effect Effects 0.000 description 3
- 238000001990 intravenous administration Methods 0.000 description 3
- XEEYBQQBJWHFJM-UHFFFAOYSA-N iron Substances [Fe] XEEYBQQBJWHFJM-UHFFFAOYSA-N 0.000 description 3
- 239000007937 lozenge Substances 0.000 description 3
- 230000036210 malignancy Effects 0.000 description 3
- 239000011159 matrix material Substances 0.000 description 3
- 108020004999 messenger RNA Proteins 0.000 description 3
- 150000002739 metals Chemical class 0.000 description 3
- 239000000178 monomer Substances 0.000 description 3
- 125000003835 nucleoside group Chemical group 0.000 description 3
- 230000005298 paramagnetic effect Effects 0.000 description 3
- 230000037361 pathway Effects 0.000 description 3
- 238000002823 phage display Methods 0.000 description 3
- 239000000546 pharmaceutical excipient Substances 0.000 description 3
- COLNVLDHVKWLRT-UHFFFAOYSA-N phenylalanine Natural products OC(=O)C(N)CC1=CC=CC=C1 COLNVLDHVKWLRT-UHFFFAOYSA-N 0.000 description 3
- 150000004713 phosphodiesters Chemical class 0.000 description 3
- 239000000843 powder Substances 0.000 description 3
- 230000035755 proliferation Effects 0.000 description 3
- 238000009877 rendering Methods 0.000 description 3
- 238000011160 research Methods 0.000 description 3
- 238000012216 screening Methods 0.000 description 3
- 238000002864 sequence alignment Methods 0.000 description 3
- 230000019491 signal transduction Effects 0.000 description 3
- 230000005477 standard model Effects 0.000 description 3
- 238000010561 standard procedure Methods 0.000 description 3
- 230000000638 stimulation Effects 0.000 description 3
- 235000000346 sugar Nutrition 0.000 description 3
- 239000006228 supernatant Substances 0.000 description 3
- 229940124597 therapeutic agent Drugs 0.000 description 3
- 229940104230 thymidine Drugs 0.000 description 3
- 238000013519 translation Methods 0.000 description 3
- 230000005909 tumor killing Effects 0.000 description 3
- QGZKDVFQNNGYKY-UHFFFAOYSA-N Ammonia Chemical compound N QGZKDVFQNNGYKY-UHFFFAOYSA-N 0.000 description 2
- 101100067974 Arabidopsis thaliana POP2 gene Proteins 0.000 description 2
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 2
- 102100038080 B-cell receptor CD22 Human genes 0.000 description 2
- 102100022005 B-lymphocyte antigen CD20 Human genes 0.000 description 2
- 102100032412 Basigin Human genes 0.000 description 2
- 102100035875 C-C chemokine receptor type 5 Human genes 0.000 description 2
- 239000002126 C01EB10 - Adenosine Substances 0.000 description 2
- 102100032937 CD40 ligand Human genes 0.000 description 2
- 108010021064 CTLA-4 Antigen Proteins 0.000 description 2
- 229940045513 CTLA4 antagonist Drugs 0.000 description 2
- 241000282472 Canis lupus familiaris Species 0.000 description 2
- 201000009030 Carcinoma Diseases 0.000 description 2
- 102000016289 Cell Adhesion Molecules Human genes 0.000 description 2
- 108010067225 Cell Adhesion Molecules Proteins 0.000 description 2
- 206010009944 Colon cancer Diseases 0.000 description 2
- 108020004635 Complementary DNA Proteins 0.000 description 2
- MIKUYHXYGGJMLM-GIMIYPNGSA-N Crotonoside Natural products C1=NC2=C(N)NC(=O)N=C2N1[C@H]1O[C@@H](CO)[C@H](O)[C@@H]1O MIKUYHXYGGJMLM-GIMIYPNGSA-N 0.000 description 2
- 102100039498 Cytotoxic T-lymphocyte protein 4 Human genes 0.000 description 2
- NYHBQMYGNKIUIF-UHFFFAOYSA-N D-guanosine Natural products C1=2NC(N)=NC(=O)C=2N=CN1C1OC(CO)C(O)C1O NYHBQMYGNKIUIF-UHFFFAOYSA-N 0.000 description 2
- 206010012335 Dependence Diseases 0.000 description 2
- SHIBSTMRCDJXLN-UHFFFAOYSA-N Digoxigenin Natural products C1CC(C2C(C3(C)CCC(O)CC3CC2)CC2O)(O)C2(C)C1C1=CC(=O)OC1 SHIBSTMRCDJXLN-UHFFFAOYSA-N 0.000 description 2
- BWGNESOTFCXPMA-UHFFFAOYSA-N Dihydrogen disulfide Chemical compound SS BWGNESOTFCXPMA-UHFFFAOYSA-N 0.000 description 2
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 description 2
- KCXVZYZYPLLWCC-UHFFFAOYSA-N EDTA Chemical compound OC(=O)CN(CC(O)=O)CCN(CC(O)=O)CC(O)=O KCXVZYZYPLLWCC-UHFFFAOYSA-N 0.000 description 2
- 108010087819 Fc receptors Proteins 0.000 description 2
- 102000009109 Fc receptors Human genes 0.000 description 2
- 238000012413 Fluorescence activated cell sorting analysis Methods 0.000 description 2
- WQZGKKKJIJFFOK-GASJEMHNSA-N Glucose Natural products OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 2
- 239000004471 Glycine Substances 0.000 description 2
- 102000003886 Glycoproteins Human genes 0.000 description 2
- 108090000288 Glycoproteins Proteins 0.000 description 2
- 239000007995 HEPES buffer Substances 0.000 description 2
- 102100022623 Hepatocyte growth factor receptor Human genes 0.000 description 2
- 101000884305 Homo sapiens B-cell receptor CD22 Proteins 0.000 description 2
- 101000897405 Homo sapiens B-lymphocyte antigen CD20 Proteins 0.000 description 2
- 101100118549 Homo sapiens EGFR gene Proteins 0.000 description 2
- 101000935040 Homo sapiens Integrin beta-2 Proteins 0.000 description 2
- 101001018097 Homo sapiens L-selectin Proteins 0.000 description 2
- 101000917826 Homo sapiens Low affinity immunoglobulin gamma Fc region receptor II-a Proteins 0.000 description 2
- 101000917824 Homo sapiens Low affinity immunoglobulin gamma Fc region receptor II-b Proteins 0.000 description 2
- 101000633784 Homo sapiens SLAM family member 7 Proteins 0.000 description 2
- 108010054477 Immunoglobulin Fab Fragments Proteins 0.000 description 2
- 102000001706 Immunoglobulin Fab Fragments Human genes 0.000 description 2
- 108010002350 Interleukin-2 Proteins 0.000 description 2
- UQSXHKLRYXJYBZ-UHFFFAOYSA-N Iron oxide Chemical compound [Fe]=O UQSXHKLRYXJYBZ-UHFFFAOYSA-N 0.000 description 2
- 208000008839 Kidney Neoplasms Diseases 0.000 description 2
- FBOZXECLQNJBKD-ZDUSSCGKSA-N L-methotrexate Chemical compound C=1N=C2N=C(N)N=C(N)C2=NC=1CN(C)C1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 FBOZXECLQNJBKD-ZDUSSCGKSA-N 0.000 description 2
- LRQKBLKVPFOOQJ-YFKPBYRVSA-N L-norleucine Chemical group CCCC[C@H]([NH3+])C([O-])=O LRQKBLKVPFOOQJ-YFKPBYRVSA-N 0.000 description 2
- 102100033467 L-selectin Human genes 0.000 description 2
- AYFVYJQAPQTCCC-GBXIJSLDSA-N L-threonine Chemical compound C[C@@H](O)[C@H](N)C(O)=O AYFVYJQAPQTCCC-GBXIJSLDSA-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
- 241000713666 Lentivirus Species 0.000 description 2
- 102100029204 Low affinity immunoglobulin gamma Fc region receptor II-a Human genes 0.000 description 2
- 206010058467 Lung neoplasm malignant Diseases 0.000 description 2
- 241000755649 Midgee Species 0.000 description 2
- 241001529936 Murinae Species 0.000 description 2
- 241000699660 Mus musculus Species 0.000 description 2
- 108010004217 Natural Cytotoxicity Triggering Receptor 1 Proteins 0.000 description 2
- 102100032870 Natural cytotoxicity triggering receptor 1 Human genes 0.000 description 2
- 208000015914 Non-Hodgkin lymphomas Diseases 0.000 description 2
- 238000012879 PET imaging Methods 0.000 description 2
- 241000276498 Pollachius virens Species 0.000 description 2
- 208000006664 Precursor Cell Lymphoblastic Leukemia-Lymphoma Diseases 0.000 description 2
- 239000004365 Protease Substances 0.000 description 2
- 239000012980 RPMI-1640 medium Substances 0.000 description 2
- 241000700159 Rattus Species 0.000 description 2
- 101710100969 Receptor tyrosine-protein kinase erbB-3 Proteins 0.000 description 2
- 102100029986 Receptor tyrosine-protein kinase erbB-3 Human genes 0.000 description 2
- 108020004511 Recombinant DNA Proteins 0.000 description 2
- 206010038389 Renal cancer Diseases 0.000 description 2
- 108020005091 Replication Origin Proteins 0.000 description 2
- 108091028664 Ribonucleotide Proteins 0.000 description 2
- 102100029198 SLAM family member 7 Human genes 0.000 description 2
- 101100123851 Saccharomyces cerevisiae (strain ATCC 204508 / S288c) HER1 gene Proteins 0.000 description 2
- 206010039491 Sarcoma Diseases 0.000 description 2
- MTCFGRXMJLQNBG-UHFFFAOYSA-N Serine Natural products OCC(N)C(O)=O MTCFGRXMJLQNBG-UHFFFAOYSA-N 0.000 description 2
- 108020004682 Single-Stranded DNA Proteins 0.000 description 2
- 241000399119 Spio Species 0.000 description 2
- AYFVYJQAPQTCCC-UHFFFAOYSA-N Threonine Natural products CC(O)C(N)C(O)=O AYFVYJQAPQTCCC-UHFFFAOYSA-N 0.000 description 2
- 239000004473 Threonine Substances 0.000 description 2
- 108700019146 Transgenes Proteins 0.000 description 2
- 208000003721 Triple Negative Breast Neoplasms 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
- ISAKRJDGNUQOIC-UHFFFAOYSA-N Uracil Chemical compound O=C1C=CNC(=O)N1 ISAKRJDGNUQOIC-UHFFFAOYSA-N 0.000 description 2
- DRTQHJPVMGBUCF-XVFCMESISA-N Uridine Chemical compound O[C@@H]1[C@H](O)[C@@H](CO)O[C@H]1N1C(=O)NC(=O)C=C1 DRTQHJPVMGBUCF-XVFCMESISA-N 0.000 description 2
- KRHYYFGTRYWZRS-BJUDXGSMSA-N ac1l2y5h Chemical compound [18FH] KRHYYFGTRYWZRS-BJUDXGSMSA-N 0.000 description 2
- 239000002253 acid Substances 0.000 description 2
- 239000013543 active substance Substances 0.000 description 2
- 229960005305 adenosine Drugs 0.000 description 2
- 239000002671 adjuvant Substances 0.000 description 2
- 230000002411 adverse Effects 0.000 description 2
- 229940124691 antibody therapeutics Drugs 0.000 description 2
- 229940049595 antibody-drug conjugate Drugs 0.000 description 2
- 210000000612 antigen-presenting cell Anatomy 0.000 description 2
- 230000006907 apoptotic process Effects 0.000 description 2
- 210000003719 b-lymphocyte Anatomy 0.000 description 2
- TZCXTZWJZNENPQ-UHFFFAOYSA-L barium sulfate Chemical compound [Ba+2].[O-]S([O-])(=O)=O TZCXTZWJZNENPQ-UHFFFAOYSA-L 0.000 description 2
- 230000008901 benefit Effects 0.000 description 2
- 230000003115 biocidal effect Effects 0.000 description 2
- 210000004369 blood Anatomy 0.000 description 2
- 239000008280 blood Substances 0.000 description 2
- 210000001124 body fluid Anatomy 0.000 description 2
- 230000009702 cancer cell proliferation Effects 0.000 description 2
- 150000001720 carbohydrates Chemical class 0.000 description 2
- 235000014633 carbohydrates Nutrition 0.000 description 2
- 229910052799 carbon Inorganic materials 0.000 description 2
- 230000015556 catabolic process Effects 0.000 description 2
- 238000004113 cell culture Methods 0.000 description 2
- 238000010367 cloning Methods 0.000 description 2
- 238000003501 co-culture Methods 0.000 description 2
- 239000003086 colorant Substances 0.000 description 2
- 230000001268 conjugating effect Effects 0.000 description 2
- 239000002872 contrast media Substances 0.000 description 2
- 238000007796 conventional method Methods 0.000 description 2
- 229910052802 copper Inorganic materials 0.000 description 2
- 238000002447 crystallographic data Methods 0.000 description 2
- 230000009089 cytolysis Effects 0.000 description 2
- 229940104302 cytosine Drugs 0.000 description 2
- 231100000433 cytotoxic Toxicity 0.000 description 2
- 238000006731 degradation reaction Methods 0.000 description 2
- 239000005547 deoxyribonucleotide Substances 0.000 description 2
- 125000002637 deoxyribonucleotide group Chemical group 0.000 description 2
- 230000029087 digestion Effects 0.000 description 2
- QONQRTHLHBTMGP-UHFFFAOYSA-N digitoxigenin Natural products CC12CCC(C3(CCC(O)CC3CC3)C)C3C11OC1CC2C1=CC(=O)OC1 QONQRTHLHBTMGP-UHFFFAOYSA-N 0.000 description 2
- SHIBSTMRCDJXLN-KCZCNTNESA-N digoxigenin Chemical compound C1([C@@H]2[C@@]3([C@@](CC2)(O)[C@H]2[C@@H]([C@@]4(C)CC[C@H](O)C[C@H]4CC2)C[C@H]3O)C)=CC(=O)OC1 SHIBSTMRCDJXLN-KCZCNTNESA-N 0.000 description 2
- 239000000539 dimer Substances 0.000 description 2
- BFMYDTVEBKDAKJ-UHFFFAOYSA-L disodium;(2',7'-dibromo-3',6'-dioxido-3-oxospiro[2-benzofuran-1,9'-xanthene]-4'-yl)mercury;hydrate Chemical compound O.[Na+].[Na+].O1C(=O)C2=CC=CC=C2C21C1=CC(Br)=C([O-])C([Hg])=C1OC1=C2C=C(Br)C([O-])=C1 BFMYDTVEBKDAKJ-UHFFFAOYSA-L 0.000 description 2
- 238000010494 dissociation reaction Methods 0.000 description 2
- 230000005593 dissociations Effects 0.000 description 2
- 239000003937 drug carrier Substances 0.000 description 2
- 238000004520 electroporation Methods 0.000 description 2
- 230000008029 eradication Effects 0.000 description 2
- 210000003527 eukaryotic cell Anatomy 0.000 description 2
- 239000007850 fluorescent dye Substances 0.000 description 2
- 239000012737 fresh medium Substances 0.000 description 2
- 239000007789 gas Substances 0.000 description 2
- 238000012239 gene modification Methods 0.000 description 2
- 230000002068 genetic effect Effects 0.000 description 2
- 230000005017 genetic modification Effects 0.000 description 2
- 235000013617 genetically modified food Nutrition 0.000 description 2
- 239000008103 glucose Substances 0.000 description 2
- 150000004676 glycans Chemical class 0.000 description 2
- UYTPUPDQBNUYGX-UHFFFAOYSA-N guanine Chemical compound O=C1NC(N)=NC2=C1N=CN2 UYTPUPDQBNUYGX-UHFFFAOYSA-N 0.000 description 2
- 229940029575 guanosine Drugs 0.000 description 2
- 210000002443 helper t lymphocyte Anatomy 0.000 description 2
- 210000005260 human cell Anatomy 0.000 description 2
- 229910052739 hydrogen Inorganic materials 0.000 description 2
- FDGQSTZJBFJUBT-UHFFFAOYSA-N hypoxanthine Chemical compound O=C1NC=NC2=C1NC=N2 FDGQSTZJBFJUBT-UHFFFAOYSA-N 0.000 description 2
- 230000000984 immunochemical effect Effects 0.000 description 2
- 238000010166 immunofluorescence Methods 0.000 description 2
- 229940072221 immunoglobulins Drugs 0.000 description 2
- 239000002596 immunotoxin Substances 0.000 description 2
- 230000008676 import Effects 0.000 description 2
- 230000000977 initiatory effect Effects 0.000 description 2
- 238000002347 injection Methods 0.000 description 2
- 239000007924 injection Substances 0.000 description 2
- 108010044426 integrins Proteins 0.000 description 2
- 102000006495 integrins Human genes 0.000 description 2
- 238000007918 intramuscular administration Methods 0.000 description 2
- 229910052742 iron Inorganic materials 0.000 description 2
- WTFXARWRTYJXII-UHFFFAOYSA-N iron(2+);iron(3+);oxygen(2-) Chemical compound [O-2].[O-2].[O-2].[O-2].[Fe+2].[Fe+3].[Fe+3] WTFXARWRTYJXII-UHFFFAOYSA-N 0.000 description 2
- 238000002955 isolation Methods 0.000 description 2
- 201000010982 kidney cancer Diseases 0.000 description 2
- 239000007788 liquid Substances 0.000 description 2
- 239000000314 lubricant Substances 0.000 description 2
- 201000005202 lung cancer Diseases 0.000 description 2
- 208000020816 lung neoplasm Diseases 0.000 description 2
- 210000004698 lymphocyte Anatomy 0.000 description 2
- 210000002540 macrophage Anatomy 0.000 description 2
- 238000002595 magnetic resonance imaging Methods 0.000 description 2
- 230000003211 malignant effect Effects 0.000 description 2
- 230000002503 metabolic effect Effects 0.000 description 2
- 229960000485 methotrexate Drugs 0.000 description 2
- 238000010369 molecular cloning Methods 0.000 description 2
- 201000000050 myeloid neoplasm Diseases 0.000 description 2
- 208000002154 non-small cell lung carcinoma Diseases 0.000 description 2
- QYSGYZVSCZSLHT-UHFFFAOYSA-N octafluoropropane Chemical compound FC(F)(F)C(F)(F)C(F)(F)F QYSGYZVSCZSLHT-UHFFFAOYSA-N 0.000 description 2
- 230000036961 partial effect Effects 0.000 description 2
- 239000002245 particle Substances 0.000 description 2
- 229960004065 perflutren Drugs 0.000 description 2
- XUYJLQHKOGNDPB-UHFFFAOYSA-N phosphonoacetic acid Chemical compound OC(=O)CP(O)(O)=O XUYJLQHKOGNDPB-UHFFFAOYSA-N 0.000 description 2
- 125000005642 phosphothioate group Chemical group 0.000 description 2
- 229920001282 polysaccharide Polymers 0.000 description 2
- 239000005017 polysaccharide Substances 0.000 description 2
- 239000013641 positive control Substances 0.000 description 2
- 230000002265 prevention Effects 0.000 description 2
- 210000001236 prokaryotic cell Anatomy 0.000 description 2
- 235000019419 proteases Nutrition 0.000 description 2
- 239000012264 purified product Substances 0.000 description 2
- 230000002285 radioactive effect Effects 0.000 description 2
- 238000012552 review Methods 0.000 description 2
- 239000002336 ribonucleotide Substances 0.000 description 2
- 125000002652 ribonucleotide group Chemical group 0.000 description 2
- 238000000926 separation method Methods 0.000 description 2
- 210000002966 serum Anatomy 0.000 description 2
- 239000011734 sodium Substances 0.000 description 2
- 239000007790 solid phase Substances 0.000 description 2
- 230000009870 specific binding Effects 0.000 description 2
- UCSJYZPVAKXKNQ-HZYVHMACSA-N streptomycin Chemical compound CN[C@H]1[C@H](O)[C@@H](O)[C@H](CO)O[C@H]1O[C@@H]1[C@](C=O)(O)[C@H](C)O[C@H]1O[C@@H]1[C@@H](NC(N)=N)[C@H](O)[C@@H](NC(N)=N)[C@H](O)[C@H]1O UCSJYZPVAKXKNQ-HZYVHMACSA-N 0.000 description 2
- 238000007920 subcutaneous administration Methods 0.000 description 2
- 239000013589 supplement Substances 0.000 description 2
- 230000004083 survival effect Effects 0.000 description 2
- 238000002560 therapeutic procedure Methods 0.000 description 2
- 229940113082 thymine Drugs 0.000 description 2
- 230000000699 topical effect Effects 0.000 description 2
- 230000001988 toxicity Effects 0.000 description 2
- 231100000419 toxicity Toxicity 0.000 description 2
- 230000002463 transducing effect Effects 0.000 description 2
- 238000003151 transfection method Methods 0.000 description 2
- 230000009466 transformation Effects 0.000 description 2
- 238000011830 transgenic mouse model Methods 0.000 description 2
- 208000022679 triple-negative breast carcinoma Diseases 0.000 description 2
- 230000004614 tumor growth Effects 0.000 description 2
- 208000029729 tumor suppressor gene on chromosome 11 Diseases 0.000 description 2
- 150000003668 tyrosines Chemical class 0.000 description 2
- 241000701161 unidentified adenovirus Species 0.000 description 2
- 229910052720 vanadium Inorganic materials 0.000 description 2
- 238000011179 visual inspection Methods 0.000 description 2
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 2
- MTCFGRXMJLQNBG-REOHCLBHSA-N (2S)-2-Amino-3-hydroxypropansäure Chemical compound OC[C@H](N)C(O)=O MTCFGRXMJLQNBG-REOHCLBHSA-N 0.000 description 1
- 102000040650 (ribonucleotides)n+m Human genes 0.000 description 1
- UKAUYVFTDYCKQA-UHFFFAOYSA-N -2-Amino-4-hydroxybutanoic acid Natural products OC(=O)C(N)CCO UKAUYVFTDYCKQA-UHFFFAOYSA-N 0.000 description 1
- UHDGCWIWMRVCDJ-UHFFFAOYSA-N 1-beta-D-Xylofuranosyl-NH-Cytosine Natural products O=C1N=C(N)C=CN1C1C(O)C(O)C(CO)O1 UHDGCWIWMRVCDJ-UHFFFAOYSA-N 0.000 description 1
- VILCJCGEZXAXTO-UHFFFAOYSA-N 2,2,2-tetramine Chemical compound NCCNCCNCCN VILCJCGEZXAXTO-UHFFFAOYSA-N 0.000 description 1
- JHALWMSZGCVVEM-UHFFFAOYSA-N 2-[4,7-bis(carboxymethyl)-1,4,7-triazonan-1-yl]acetic acid Chemical compound OC(=O)CN1CCN(CC(O)=O)CCN(CC(O)=O)CC1 JHALWMSZGCVVEM-UHFFFAOYSA-N 0.000 description 1
- AOYNUTHNTBLRMT-SLPGGIOYSA-N 2-deoxy-2-fluoro-aldehydo-D-glucose Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)[C@@H](F)C=O AOYNUTHNTBLRMT-SLPGGIOYSA-N 0.000 description 1
- ASJSAQIRZKANQN-CRCLSJGQSA-N 2-deoxy-D-ribose Chemical compound OC[C@@H](O)[C@@H](O)CC=O ASJSAQIRZKANQN-CRCLSJGQSA-N 0.000 description 1
- UAIUNKRWKOVEES-UHFFFAOYSA-N 3,3',5,5'-tetramethylbenzidine Chemical compound CC1=C(N)C(C)=CC(C=2C=C(C)C(N)=C(C)C=2)=C1 UAIUNKRWKOVEES-UHFFFAOYSA-N 0.000 description 1
- TVZGACDUOSZQKY-LBPRGKRZSA-N 4-aminofolic acid Chemical compound C1=NC2=NC(N)=NC(N)=C2N=C1CNC1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 TVZGACDUOSZQKY-LBPRGKRZSA-N 0.000 description 1
- ZAYHVCMSTBRABG-UHFFFAOYSA-N 5-Methylcytidine Natural products O=C1N=C(N)C(C)=CN1C1C(O)C(O)C(CO)O1 ZAYHVCMSTBRABG-UHFFFAOYSA-N 0.000 description 1
- ZAYHVCMSTBRABG-JXOAFFINSA-N 5-methylcytidine Chemical compound O=C1N=C(N)C(C)=CN1[C@H]1[C@H](O)[C@H](O)[C@@H](CO)O1 ZAYHVCMSTBRABG-JXOAFFINSA-N 0.000 description 1
- 102100023990 60S ribosomal protein L17 Human genes 0.000 description 1
- 102100031585 ADP-ribosyl cyclase/cyclic ADP-ribose hydrolase 1 Human genes 0.000 description 1
- 206010000060 Abdominal distension Diseases 0.000 description 1
- QTBSBXVTEAMEQO-UHFFFAOYSA-M Acetate Chemical compound CC([O-])=O QTBSBXVTEAMEQO-UHFFFAOYSA-M 0.000 description 1
- 208000024893 Acute lymphoblastic leukemia Diseases 0.000 description 1
- 208000014697 Acute lymphocytic leukaemia Diseases 0.000 description 1
- 208000031261 Acute myeloid leukaemia Diseases 0.000 description 1
- 229930024421 Adenine Natural products 0.000 description 1
- GFFGJBXGBJISGV-UHFFFAOYSA-N Adenine Chemical compound NC1=NC=NC2=C1N=CN2 GFFGJBXGBJISGV-UHFFFAOYSA-N 0.000 description 1
- 108010088751 Albumins Proteins 0.000 description 1
- 102000009027 Albumins Human genes 0.000 description 1
- 108700028369 Alleles Proteins 0.000 description 1
- 102100035248 Alpha-(1,3)-fucosyltransferase 4 Human genes 0.000 description 1
- QGZKDVFQNNGYKY-UHFFFAOYSA-O Ammonium Chemical compound [NH4+] QGZKDVFQNNGYKY-UHFFFAOYSA-O 0.000 description 1
- 229920000856 Amylose Polymers 0.000 description 1
- 108010032595 Antibody Binding Sites Proteins 0.000 description 1
- 239000004475 Arginine Substances 0.000 description 1
- DCXYFEDJOCDNAF-UHFFFAOYSA-N Asparagine Natural products OC(=O)C(N)CC(N)=O DCXYFEDJOCDNAF-UHFFFAOYSA-N 0.000 description 1
- 241001367053 Autographa gamma Species 0.000 description 1
- 238000012935 Averaging Methods 0.000 description 1
- 208000003950 B-cell lymphoma Diseases 0.000 description 1
- 101710117995 B-lymphocyte antigen CD19 Proteins 0.000 description 1
- 241000894006 Bacteria Species 0.000 description 1
- 108010064528 Basigin Proteins 0.000 description 1
- 206010005003 Bladder cancer Diseases 0.000 description 1
- 206010005949 Bone cancer Diseases 0.000 description 1
- 208000018084 Bone neoplasm Diseases 0.000 description 1
- 208000011691 Burkitt lymphomas Diseases 0.000 description 1
- 101710149870 C-C chemokine receptor type 5 Proteins 0.000 description 1
- 102100024217 CAMPATH-1 antigen Human genes 0.000 description 1
- 108010005327 CD19-specific chimeric antigen receptor Proteins 0.000 description 1
- 108010029697 CD40 Ligand Proteins 0.000 description 1
- 101150013553 CD40 gene Proteins 0.000 description 1
- 102100032912 CD44 antigen Human genes 0.000 description 1
- 108010065524 CD52 Antigen Proteins 0.000 description 1
- 108091008048 CMVpp65 Proteins 0.000 description 1
- OYPRJOBELJOOCE-UHFFFAOYSA-N Calcium Chemical compound [Ca] OYPRJOBELJOOCE-UHFFFAOYSA-N 0.000 description 1
- OKTJSMMVPCPJKN-UHFFFAOYSA-N Carbon Chemical compound [C] OKTJSMMVPCPJKN-UHFFFAOYSA-N 0.000 description 1
- 241000700199 Cavia porcellus Species 0.000 description 1
- 102000000844 Cell Surface Receptors Human genes 0.000 description 1
- 108010001857 Cell Surface Receptors Proteins 0.000 description 1
- 241000282693 Cercopithecidae Species 0.000 description 1
- 229910052684 Cerium Inorganic materials 0.000 description 1
- 241000282994 Cervidae Species 0.000 description 1
- 206010008342 Cervix carcinoma Diseases 0.000 description 1
- 241001227713 Chiron Species 0.000 description 1
- 108091062157 Cis-regulatory element Proteins 0.000 description 1
- 208000001333 Colorectal Neoplasms Diseases 0.000 description 1
- 102000000989 Complement System Proteins Human genes 0.000 description 1
- 108010069112 Complement System Proteins Proteins 0.000 description 1
- 206010010144 Completed suicide Diseases 0.000 description 1
- RYGMFSIKBFXOCR-UHFFFAOYSA-N Copper Chemical compound [Cu] RYGMFSIKBFXOCR-UHFFFAOYSA-N 0.000 description 1
- UHDGCWIWMRVCDJ-PSQAKQOGSA-N Cytidine Natural products O=C1N=C(N)C=CN1[C@@H]1[C@@H](O)[C@@H](O)[C@H](CO)O1 UHDGCWIWMRVCDJ-PSQAKQOGSA-N 0.000 description 1
- 241000701022 Cytomegalovirus Species 0.000 description 1
- HMFHBZSHGGEWLO-SOOFDHNKSA-N D-ribofuranose Chemical compound OC[C@H]1OC(O)[C@H](O)[C@@H]1O HMFHBZSHGGEWLO-SOOFDHNKSA-N 0.000 description 1
- 238000000018 DNA microarray Methods 0.000 description 1
- 241000450599 DNA viruses Species 0.000 description 1
- 241000702421 Dependoparvovirus Species 0.000 description 1
- FEWJPZIEWOKRBE-JCYAYHJZSA-N Dextrotartaric acid Chemical compound OC(=O)[C@H](O)[C@@H](O)C(O)=O FEWJPZIEWOKRBE-JCYAYHJZSA-N 0.000 description 1
- 206010061818 Disease progression Diseases 0.000 description 1
- 229910052692 Dysprosium Inorganic materials 0.000 description 1
- 102100025137 Early activation antigen CD69 Human genes 0.000 description 1
- 101800003838 Epidermal growth factor Proteins 0.000 description 1
- YQYJSBFKSSDGFO-UHFFFAOYSA-N Epihygromycin Natural products OC1C(O)C(C(=O)C)OC1OC(C(=C1)O)=CC=C1C=C(C)C(=O)NC1C(O)C(O)C2OCOC2C1O YQYJSBFKSSDGFO-UHFFFAOYSA-N 0.000 description 1
- 102000018651 Epithelial Cell Adhesion Molecule Human genes 0.000 description 1
- 108010066687 Epithelial Cell Adhesion Molecule Proteins 0.000 description 1
- 241000214054 Equine rhinitis A virus Species 0.000 description 1
- 229910052691 Erbium Inorganic materials 0.000 description 1
- 208000000461 Esophageal Neoplasms Diseases 0.000 description 1
- 241000206602 Eukaryota Species 0.000 description 1
- 229910052693 Europium Inorganic materials 0.000 description 1
- 208000010201 Exanthema Diseases 0.000 description 1
- 108700024394 Exon Proteins 0.000 description 1
- 241000282326 Felis catus Species 0.000 description 1
- 241000710198 Foot-and-mouth disease virus Species 0.000 description 1
- GYHNNYVSQQEPJS-UHFFFAOYSA-N Gallium Chemical compound [Ga] GYHNNYVSQQEPJS-UHFFFAOYSA-N 0.000 description 1
- 108010010803 Gelatin Proteins 0.000 description 1
- 108700028146 Genetic Enhancer Elements Proteins 0.000 description 1
- 208000032612 Glial tumor Diseases 0.000 description 1
- 206010018338 Glioma Diseases 0.000 description 1
- 102100041003 Glutamate carboxypeptidase 2 Human genes 0.000 description 1
- 102000004269 Granulocyte Colony-Stimulating Factor Human genes 0.000 description 1
- 108010017080 Granulocyte Colony-Stimulating Factor Proteins 0.000 description 1
- 102100039622 Granulocyte colony-stimulating factor receptor Human genes 0.000 description 1
- 108020005004 Guide RNA Proteins 0.000 description 1
- 102100030595 HLA class II histocompatibility antigen gamma chain Human genes 0.000 description 1
- 102000006354 HLA-DR Antigens Human genes 0.000 description 1
- 108010058597 HLA-DR Antigens Proteins 0.000 description 1
- 102100031573 Hematopoietic progenitor cell antigen CD34 Human genes 0.000 description 1
- 241000238631 Hexapoda Species 0.000 description 1
- 208000017604 Hodgkin disease Diseases 0.000 description 1
- 208000021519 Hodgkin lymphoma Diseases 0.000 description 1
- 208000010747 Hodgkins lymphoma Diseases 0.000 description 1
- 229910052689 Holmium Inorganic materials 0.000 description 1
- 101000777636 Homo sapiens ADP-ribosyl cyclase/cyclic ADP-ribose hydrolase 1 Proteins 0.000 description 1
- 101001022185 Homo sapiens Alpha-(1,3)-fucosyltransferase 4 Proteins 0.000 description 1
- 101000798441 Homo sapiens Basigin Proteins 0.000 description 1
- 101000868273 Homo sapiens CD44 antigen Proteins 0.000 description 1
- 101000914324 Homo sapiens Carcinoembryonic antigen-related cell adhesion molecule 5 Proteins 0.000 description 1
- 101000914321 Homo sapiens Carcinoembryonic antigen-related cell adhesion molecule 7 Proteins 0.000 description 1
- 101000934374 Homo sapiens Early activation antigen CD69 Proteins 0.000 description 1
- 101000892862 Homo sapiens Glutamate carboxypeptidase 2 Proteins 0.000 description 1
- 101000746364 Homo sapiens Granulocyte colony-stimulating factor receptor Proteins 0.000 description 1
- 101001082627 Homo sapiens HLA class II histocompatibility antigen gamma chain Proteins 0.000 description 1
- 101000777663 Homo sapiens Hematopoietic progenitor cell antigen CD34 Proteins 0.000 description 1
- 101000972946 Homo sapiens Hepatocyte growth factor receptor Proteins 0.000 description 1
- 101001076642 Homo sapiens Inosine-5'-monophosphate dehydrogenase 2 Proteins 0.000 description 1
- 101001078143 Homo sapiens Integrin alpha-IIb Proteins 0.000 description 1
- 101001046677 Homo sapiens Integrin alpha-V Proteins 0.000 description 1
- 101001057504 Homo sapiens Interferon-stimulated gene 20 kDa protein Proteins 0.000 description 1
- 101001055157 Homo sapiens Interleukin-15 Proteins 0.000 description 1
- 101001055144 Homo sapiens Interleukin-2 receptor subunit alpha Proteins 0.000 description 1
- 101000960936 Homo sapiens Interleukin-5 receptor subunit alpha Proteins 0.000 description 1
- 101000878605 Homo sapiens Low affinity immunoglobulin epsilon Fc receptor Proteins 0.000 description 1
- 101001133056 Homo sapiens Mucin-1 Proteins 0.000 description 1
- 101000623901 Homo sapiens Mucin-16 Proteins 0.000 description 1
- 101000934338 Homo sapiens Myeloid cell surface antigen CD33 Proteins 0.000 description 1
- 101000617725 Homo sapiens Pregnancy-specific beta-1-glycoprotein 2 Proteins 0.000 description 1
- 101000914484 Homo sapiens T-lymphocyte activation antigen CD80 Proteins 0.000 description 1
- 101000904724 Homo sapiens Transmembrane glycoprotein NMB Proteins 0.000 description 1
- 101000801433 Homo sapiens Trophoblast glycoprotein Proteins 0.000 description 1
- 101000610605 Homo sapiens Tumor necrosis factor receptor superfamily member 10A Proteins 0.000 description 1
- 101000610604 Homo sapiens Tumor necrosis factor receptor superfamily member 10B Proteins 0.000 description 1
- 101000851376 Homo sapiens Tumor necrosis factor receptor superfamily member 8 Proteins 0.000 description 1
- VEXZGXHMUGYJMC-UHFFFAOYSA-N Hydrochloric acid Chemical compound Cl VEXZGXHMUGYJMC-UHFFFAOYSA-N 0.000 description 1
- CPELXLSAUQHCOX-UHFFFAOYSA-N Hydrogen bromide Chemical compound Br CPELXLSAUQHCOX-UHFFFAOYSA-N 0.000 description 1
- PMMYEEVYMWASQN-DMTCNVIQSA-N Hydroxyproline Chemical compound O[C@H]1CN[C@H](C(O)=O)C1 PMMYEEVYMWASQN-DMTCNVIQSA-N 0.000 description 1
- UGQMRVRMYYASKQ-UHFFFAOYSA-N Hypoxanthine nucleoside Natural products OC1C(O)C(CO)OC1N1C(NC=NC2=O)=C2N=C1 UGQMRVRMYYASKQ-UHFFFAOYSA-N 0.000 description 1
- 102000009438 IgE Receptors Human genes 0.000 description 1
- 108010073816 IgE Receptors Proteins 0.000 description 1
- DGAQECJNVWCQMB-PUAWFVPOSA-M Ilexoside XXIX Chemical compound C[C@@H]1CC[C@@]2(CC[C@@]3(C(=CC[C@H]4[C@]3(CC[C@@H]5[C@@]4(CC[C@@H](C5(C)C)OS(=O)(=O)[O-])C)C)[C@@H]2[C@]1(C)O)C)C(=O)O[C@H]6[C@@H]([C@H]([C@@H]([C@H](O6)CO)O)O)O.[Na+] DGAQECJNVWCQMB-PUAWFVPOSA-M 0.000 description 1
- 108010067060 Immunoglobulin Variable Region Proteins 0.000 description 1
- 102000017727 Immunoglobulin Variable Region Human genes 0.000 description 1
- 229930010555 Inosine Natural products 0.000 description 1
- UGQMRVRMYYASKQ-KQYNXXCUSA-N Inosine Chemical compound O[C@@H]1[C@H](O)[C@@H](CO)O[C@H]1N1C2=NC=NC(O)=C2N=C1 UGQMRVRMYYASKQ-KQYNXXCUSA-N 0.000 description 1
- 102100025891 Inosine-5'-monophosphate dehydrogenase 2 Human genes 0.000 description 1
- 108090000723 Insulin-Like Growth Factor I Proteins 0.000 description 1
- 102100025306 Integrin alpha-IIb Human genes 0.000 description 1
- 102100022337 Integrin alpha-V Human genes 0.000 description 1
- 108010064593 Intercellular Adhesion Molecule-1 Proteins 0.000 description 1
- 102100037877 Intercellular adhesion molecule 1 Human genes 0.000 description 1
- 102000001617 Interferon Receptors Human genes 0.000 description 1
- 108010054267 Interferon Receptors Proteins 0.000 description 1
- 102100027268 Interferon-stimulated gene 20 kDa protein Human genes 0.000 description 1
- 102100039881 Interleukin-5 receptor subunit alpha Human genes 0.000 description 1
- 108010038501 Interleukin-6 Receptors Proteins 0.000 description 1
- 102000010781 Interleukin-6 Receptors Human genes 0.000 description 1
- AMDBBAQNWSUWGN-UHFFFAOYSA-N Ioversol Chemical compound OCCN(C(=O)CO)C1=C(I)C(C(=O)NCC(O)CO)=C(I)C(C(=O)NCC(O)CO)=C1I AMDBBAQNWSUWGN-UHFFFAOYSA-N 0.000 description 1
- 241001197082 Knodus beta Species 0.000 description 1
- 101150062031 L gene Proteins 0.000 description 1
- DCXYFEDJOCDNAF-REOHCLBHSA-N L-asparagine Chemical compound OC(=O)[C@@H](N)CC(N)=O DCXYFEDJOCDNAF-REOHCLBHSA-N 0.000 description 1
- CKLJMWTZIZZHCS-REOHCLBHSA-N L-aspartic acid Chemical compound OC(=O)[C@@H](N)CC(O)=O CKLJMWTZIZZHCS-REOHCLBHSA-N 0.000 description 1
- UKAUYVFTDYCKQA-VKHMYHEASA-N L-homoserine Chemical group OC(=O)[C@@H](N)CCO UKAUYVFTDYCKQA-VKHMYHEASA-N 0.000 description 1
- AGPKZVBTJJNPAG-WHFBIAKZSA-N L-isoleucine Chemical compound CC[C@H](C)[C@H](N)C(O)=O AGPKZVBTJJNPAG-WHFBIAKZSA-N 0.000 description 1
- ROHFNLRQFUQHCH-YFKPBYRVSA-N L-leucine Chemical compound CC(C)C[C@H](N)C(O)=O ROHFNLRQFUQHCH-YFKPBYRVSA-N 0.000 description 1
- QEFRNWWLZKMPFJ-ZXPFJRLXSA-N L-methionine (R)-S-oxide Chemical group C[S@@](=O)CC[C@H]([NH3+])C([O-])=O QEFRNWWLZKMPFJ-ZXPFJRLXSA-N 0.000 description 1
- QEFRNWWLZKMPFJ-UHFFFAOYSA-N L-methionine sulphoxide Chemical group CS(=O)CCC(N)C(O)=O QEFRNWWLZKMPFJ-UHFFFAOYSA-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
- GUBGYTABKSRVRQ-QKKXKWKRSA-N Lactose Natural products OC[C@H]1O[C@@H](O[C@H]2[C@H](O)[C@@H](O)C(O)O[C@@H]2CO)[C@H](O)[C@@H](O)[C@H]1O GUBGYTABKSRVRQ-QKKXKWKRSA-N 0.000 description 1
- 241000270322 Lepidosauria Species 0.000 description 1
- ROHFNLRQFUQHCH-UHFFFAOYSA-N Leucine Natural products CC(C)CC(N)C(O)=O ROHFNLRQFUQHCH-UHFFFAOYSA-N 0.000 description 1
- 102100038007 Low affinity immunoglobulin epsilon Fc receptor Human genes 0.000 description 1
- 229910052765 Lutetium Inorganic materials 0.000 description 1
- 108010064548 Lymphocyte Function-Associated Antigen-1 Proteins 0.000 description 1
- 208000031422 Lymphocytic Chronic B-Cell Leukemia Diseases 0.000 description 1
- 239000004472 Lysine Substances 0.000 description 1
- KDXKERNSBIXSRK-UHFFFAOYSA-N Lysine Natural products NCCCCC(N)C(O)=O KDXKERNSBIXSRK-UHFFFAOYSA-N 0.000 description 1
- FYYHWMGAXLPEAU-UHFFFAOYSA-N Magnesium Chemical compound [Mg] FYYHWMGAXLPEAU-UHFFFAOYSA-N 0.000 description 1
- 102000018697 Membrane Proteins Human genes 0.000 description 1
- 108010052285 Membrane Proteins Proteins 0.000 description 1
- AFVFQIVMOAPDHO-UHFFFAOYSA-N Methanesulfonic acid Chemical compound CS(O)(=O)=O AFVFQIVMOAPDHO-UHFFFAOYSA-N 0.000 description 1
- 102100034256 Mucin-1 Human genes 0.000 description 1
- 102100023123 Mucin-16 Human genes 0.000 description 1
- 208000034578 Multiple myelomas Diseases 0.000 description 1
- 101001033610 Mycobacterium tuberculosis (strain ATCC 25618 / H37Rv) Inosine-5'-monophosphate dehydrogenase Proteins 0.000 description 1
- 102100025243 Myeloid cell surface antigen CD33 Human genes 0.000 description 1
- QPCDCPDFJACHGM-UHFFFAOYSA-N N,N-bis{2-[bis(carboxymethyl)amino]ethyl}glycine Chemical compound OC(=O)CN(CC(O)=O)CCN(CC(=O)O)CCN(CC(O)=O)CC(O)=O QPCDCPDFJACHGM-UHFFFAOYSA-N 0.000 description 1
- 229930182474 N-glycoside Natural products 0.000 description 1
- 229910052779 Neodymium Inorganic materials 0.000 description 1
- 108010032605 Nerve Growth Factor Receptors Proteins 0.000 description 1
- 102000007339 Nerve Growth Factor Receptors Human genes 0.000 description 1
- QJGQUHMNIGDVPM-BJUDXGSMSA-N Nitrogen-13 Chemical compound [13N] QJGQUHMNIGDVPM-BJUDXGSMSA-N 0.000 description 1
- 241000283973 Oryctolagus cuniculus Species 0.000 description 1
- MUBZPKHOEPUJKR-UHFFFAOYSA-N Oxalic acid Chemical compound OC(=O)C(O)=O MUBZPKHOEPUJKR-UHFFFAOYSA-N 0.000 description 1
- LYNKVJADAPZJIK-UHFFFAOYSA-H P([O-])([O-])=O.[B+3].P([O-])([O-])=O.P([O-])([O-])=O.[B+3] Chemical compound P([O-])([O-])=O.[B+3].P([O-])([O-])=O.P([O-])([O-])=O.[B+3] LYNKVJADAPZJIK-UHFFFAOYSA-H 0.000 description 1
- 108091008606 PDGF receptors Proteins 0.000 description 1
- 229910019142 PO4 Inorganic materials 0.000 description 1
- 241000282577 Pan troglodytes Species 0.000 description 1
- 206010061902 Pancreatic neoplasm Diseases 0.000 description 1
- 241001494479 Pecora Species 0.000 description 1
- 229930182555 Penicillin Natural products 0.000 description 1
- JGSARLDLIJGVTE-MBNYWOFBSA-N Penicillin G Chemical compound N([C@H]1[C@H]2SC([C@@H](N2C1=O)C(O)=O)(C)C)C(=O)CC1=CC=CC=C1 JGSARLDLIJGVTE-MBNYWOFBSA-N 0.000 description 1
- 102000057297 Pepsin A Human genes 0.000 description 1
- 108090000284 Pepsin A Proteins 0.000 description 1
- 108010077524 Peptide Elongation Factor 1 Proteins 0.000 description 1
- 108091093037 Peptide nucleic acid Proteins 0.000 description 1
- 229940122907 Phosphatase inhibitor Drugs 0.000 description 1
- 102000011653 Platelet-Derived Growth Factor Receptors Human genes 0.000 description 1
- 229920002535 Polyethylene Glycol 1500 Polymers 0.000 description 1
- 108010039918 Polylysine Proteins 0.000 description 1
- 229920001213 Polysorbate 20 Polymers 0.000 description 1
- 241001672814 Porcine teschovirus 1 Species 0.000 description 1
- ZLMJMSJWJFRBEC-UHFFFAOYSA-N Potassium Chemical compound [K] ZLMJMSJWJFRBEC-UHFFFAOYSA-N 0.000 description 1
- 229910052777 Praseodymium Inorganic materials 0.000 description 1
- 102100022019 Pregnancy-specific beta-1-glycoprotein 2 Human genes 0.000 description 1
- 241000288906 Primates Species 0.000 description 1
- 206010036790 Productive cough Diseases 0.000 description 1
- 101710089372 Programmed cell death protein 1 Proteins 0.000 description 1
- 206010060862 Prostate cancer Diseases 0.000 description 1
- 208000000236 Prostatic Neoplasms Diseases 0.000 description 1
- 108010029485 Protein Isoforms Proteins 0.000 description 1
- 102000001708 Protein Isoforms Human genes 0.000 description 1
- 108010089836 Proto-Oncogene Proteins c-met Proteins 0.000 description 1
- 229930185560 Pseudouridine Natural products 0.000 description 1
- PTJWIQPHWPFNBW-UHFFFAOYSA-N Pseudouridine C Natural products OC1C(O)C(CO)OC1C1=CNC(=O)NC1=O PTJWIQPHWPFNBW-UHFFFAOYSA-N 0.000 description 1
- 102100029981 Receptor tyrosine-protein kinase erbB-4 Human genes 0.000 description 1
- 101710100963 Receptor tyrosine-protein kinase erbB-4 Proteins 0.000 description 1
- 108700008625 Reporter Genes Proteins 0.000 description 1
- 208000007660 Residual Neoplasm Diseases 0.000 description 1
- PYMYPHUHKUWMLA-LMVFSUKVSA-N Ribose Natural products OC[C@@H](O)[C@@H](O)[C@@H](O)C=O PYMYPHUHKUWMLA-LMVFSUKVSA-N 0.000 description 1
- IGLNJRXAVVLDKE-OIOBTWANSA-N Rubidium-82 Chemical compound [82Rb] IGLNJRXAVVLDKE-OIOBTWANSA-N 0.000 description 1
- 229910052772 Samarium Inorganic materials 0.000 description 1
- 108020004459 Small interfering RNA Proteins 0.000 description 1
- 102000013275 Somatomedins Human genes 0.000 description 1
- 241000256248 Spodoptera Species 0.000 description 1
- 229920002472 Starch Polymers 0.000 description 1
- 208000005718 Stomach Neoplasms Diseases 0.000 description 1
- 108010090804 Streptavidin Proteins 0.000 description 1
- 229930006000 Sucrose Natural products 0.000 description 1
- CZMRCDWAGMRECN-UGDNZRGBSA-N Sucrose Chemical compound O[C@H]1[C@H](O)[C@@H](CO)O[C@@]1(CO)O[C@@H]1[C@H](O)[C@@H](O)[C@H](O)[C@@H](CO)O1 CZMRCDWAGMRECN-UGDNZRGBSA-N 0.000 description 1
- NINIDFKCEFEMDL-UHFFFAOYSA-N Sulfur Chemical compound [S] NINIDFKCEFEMDL-UHFFFAOYSA-N 0.000 description 1
- 239000012505 Superdex™ Substances 0.000 description 1
- 102100027222 T-lymphocyte activation antigen CD80 Human genes 0.000 description 1
- 229910052771 Terbium Inorganic materials 0.000 description 1
- WDLRUFUQRNWCPK-UHFFFAOYSA-N Tetraxetan Chemical compound OC(=O)CN1CCN(CC(O)=O)CCN(CC(O)=O)CCN(CC(O)=O)CC1 WDLRUFUQRNWCPK-UHFFFAOYSA-N 0.000 description 1
- 238000012338 Therapeutic targeting Methods 0.000 description 1
- 241001648840 Thosea asigna virus Species 0.000 description 1
- 229910052775 Thulium Inorganic materials 0.000 description 1
- 230000010632 Transcription Factor Activity Effects 0.000 description 1
- 102100037116 Transcription elongation factor 1 homolog Human genes 0.000 description 1
- 102100023935 Transmembrane glycoprotein NMB Human genes 0.000 description 1
- 108060008683 Tumor Necrosis Factor Receptor Proteins 0.000 description 1
- 102100040113 Tumor necrosis factor receptor superfamily member 10A Human genes 0.000 description 1
- 102100040112 Tumor necrosis factor receptor superfamily member 10B Human genes 0.000 description 1
- 102100040245 Tumor necrosis factor receptor superfamily member 5 Human genes 0.000 description 1
- 102100036857 Tumor necrosis factor receptor superfamily member 8 Human genes 0.000 description 1
- 206010053613 Type IV hypersensitivity reaction Diseases 0.000 description 1
- 208000006105 Uterine Cervical Neoplasms Diseases 0.000 description 1
- 108091008605 VEGF receptors Proteins 0.000 description 1
- 102000009484 Vascular Endothelial Growth Factor Receptors Human genes 0.000 description 1
- 108010034265 Vascular Endothelial Growth Factor Receptors Proteins 0.000 description 1
- 102100033177 Vascular endothelial growth factor receptor 2 Human genes 0.000 description 1
- 108020005202 Viral DNA Proteins 0.000 description 1
- 229910052769 Ytterbium Inorganic materials 0.000 description 1
- JLCPHMBAVCMARE-UHFFFAOYSA-N [3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-hydroxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methyl [5-(6-aminopurin-9-yl)-2-(hydroxymethyl)oxolan-3-yl] hydrogen phosphate Polymers Cc1cn(C2CC(OP(O)(=O)OCC3OC(CC3OP(O)(=O)OCC3OC(CC3O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c3nc(N)[nH]c4=O)C(COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3CO)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cc(C)c(=O)[nH]c3=O)n3cc(C)c(=O)[nH]c3=O)n3ccc(N)nc3=O)n3cc(C)c(=O)[nH]c3=O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)O2)c(=O)[nH]c1=O JLCPHMBAVCMARE-UHFFFAOYSA-N 0.000 description 1
- ZUPXXZAVUHFCNV-UHFFFAOYSA-N [[5-(6-aminopurin-9-yl)-3,4-dihydroxyoxolan-2-yl]methoxy-hydroxyphosphoryl] [5-(3-carbamoyl-4h-pyridin-1-yl)-3,4-dihydroxyoxolan-2-yl]methyl hydrogen phosphate;potassium Chemical compound [K].C1=CCC(C(=O)N)=CN1C1C(O)C(O)C(COP(O)(=O)OP(O)(=O)OCC2C(C(O)C(O2)N2C3=NC=NC(N)=C3N=C2)O)O1 ZUPXXZAVUHFCNV-UHFFFAOYSA-N 0.000 description 1
- 238000002679 ablation Methods 0.000 description 1
- 238000010521 absorption reaction Methods 0.000 description 1
- 238000009825 accumulation Methods 0.000 description 1
- 150000007513 acids Chemical class 0.000 description 1
- 239000004480 active ingredient Substances 0.000 description 1
- 230000033289 adaptive immune response Effects 0.000 description 1
- 238000011374 additional therapy Methods 0.000 description 1
- 229960000643 adenine Drugs 0.000 description 1
- 208000009956 adenocarcinoma Diseases 0.000 description 1
- 239000000443 aerosol Substances 0.000 description 1
- 230000002776 aggregation Effects 0.000 description 1
- 238000004220 aggregation Methods 0.000 description 1
- 150000001298 alcohols Chemical class 0.000 description 1
- PPQRONHOSHZGFQ-LMVFSUKVSA-N aldehydo-D-ribose 5-phosphate Chemical group OP(=O)(O)OC[C@@H](O)[C@@H](O)[C@@H](O)C=O PPQRONHOSHZGFQ-LMVFSUKVSA-N 0.000 description 1
- HMFHBZSHGGEWLO-UHFFFAOYSA-N alpha-D-Furanose-Ribose Natural products OCC1OC(O)C(O)C1O HMFHBZSHGGEWLO-UHFFFAOYSA-N 0.000 description 1
- WQZGKKKJIJFFOK-PHYPRBDBSA-N alpha-D-galactose Chemical compound OC[C@H]1O[C@H](O)[C@H](O)[C@@H](O)[C@H]1O WQZGKKKJIJFFOK-PHYPRBDBSA-N 0.000 description 1
- YVPYQUNUQOZFHG-UHFFFAOYSA-N amidotrizoic acid Chemical compound CC(=O)NC1=C(I)C(NC(C)=O)=C(I)C(C(O)=O)=C1I YVPYQUNUQOZFHG-UHFFFAOYSA-N 0.000 description 1
- 125000003277 amino group Chemical group 0.000 description 1
- 229960003896 aminopterin Drugs 0.000 description 1
- 229910021529 ammonia Inorganic materials 0.000 description 1
- 238000004873 anchoring Methods 0.000 description 1
- 125000000129 anionic group Chemical group 0.000 description 1
- 230000000118 anti-neoplastic effect Effects 0.000 description 1
- 239000000611 antibody drug conjugate Substances 0.000 description 1
- 230000000890 antigenic effect Effects 0.000 description 1
- 239000007864 aqueous solution Substances 0.000 description 1
- ODKSFYDXXFIFQN-UHFFFAOYSA-N arginine Natural products OC(=O)C(N)CCCNC(N)=N ODKSFYDXXFIFQN-UHFFFAOYSA-N 0.000 description 1
- 210000002565 arteriole Anatomy 0.000 description 1
- 125000003118 aryl group Chemical group 0.000 description 1
- 235000009582 asparagine Nutrition 0.000 description 1
- 229960001230 asparagine Drugs 0.000 description 1
- 235000003704 aspartic acid Nutrition 0.000 description 1
- QVGXLLKOCUKJST-UHFFFAOYSA-N atomic oxygen Chemical compound [O] QVGXLLKOCUKJST-UHFFFAOYSA-N 0.000 description 1
- 230000035578 autophosphorylation Effects 0.000 description 1
- 238000011888 autopsy Methods 0.000 description 1
- 239000012752 auxiliary agent Substances 0.000 description 1
- DRTQHJPVMGBUCF-PSQAKQOGSA-N beta-L-uridine Natural products O[C@H]1[C@@H](O)[C@H](CO)O[C@@H]1N1C(=O)NC(=O)C=C1 DRTQHJPVMGBUCF-PSQAKQOGSA-N 0.000 description 1
- WGDUUQDYDIIBKT-UHFFFAOYSA-N beta-Pseudouridine Natural products OC1OC(CN2C=CC(=O)NC2=O)C(O)C1O WGDUUQDYDIIBKT-UHFFFAOYSA-N 0.000 description 1
- OQFSQFPPLPISGP-UHFFFAOYSA-N beta-carboxyaspartic acid Natural products OC(=O)C(N)C(C(O)=O)C(O)=O OQFSQFPPLPISGP-UHFFFAOYSA-N 0.000 description 1
- 238000001574 biopsy Methods 0.000 description 1
- 230000006287 biotinylation Effects 0.000 description 1
- 238000007413 biotinylation Methods 0.000 description 1
- 201000001531 bladder carcinoma Diseases 0.000 description 1
- 210000001772 blood platelet Anatomy 0.000 description 1
- 210000001185 bone marrow Anatomy 0.000 description 1
- 210000002798 bone marrow cell Anatomy 0.000 description 1
- 210000004556 brain Anatomy 0.000 description 1
- 210000000481 breast Anatomy 0.000 description 1
- 230000003139 buffering effect Effects 0.000 description 1
- 239000011575 calcium Substances 0.000 description 1
- 229910052791 calcium Inorganic materials 0.000 description 1
- 239000001506 calcium phosphate Substances 0.000 description 1
- 229910000389 calcium phosphate Inorganic materials 0.000 description 1
- 235000011010 calcium phosphates Nutrition 0.000 description 1
- BPKIGYQJPYCAOW-FFJTTWKXSA-I calcium;potassium;disodium;(2s)-2-hydroxypropanoate;dichloride;dihydroxide;hydrate Chemical compound O.[OH-].[OH-].[Na+].[Na+].[Cl-].[Cl-].[K+].[Ca+2].C[C@H](O)C([O-])=O BPKIGYQJPYCAOW-FFJTTWKXSA-I 0.000 description 1
- 238000004364 calculation method Methods 0.000 description 1
- 208000035269 cancer or benign tumor Diseases 0.000 description 1
- 229910002091 carbon monoxide Inorganic materials 0.000 description 1
- OKTJSMMVPCPJKN-BJUDXGSMSA-N carbon-11 Chemical compound [11C] OKTJSMMVPCPJKN-BJUDXGSMSA-N 0.000 description 1
- 239000011545 carbonate/bicarbonate buffer Substances 0.000 description 1
- 125000003178 carboxy group Chemical group [H]OC(*)=O 0.000 description 1
- 230000030833 cell death Effects 0.000 description 1
- 230000032823 cell division Effects 0.000 description 1
- 230000003915 cell function Effects 0.000 description 1
- 230000007910 cell fusion Effects 0.000 description 1
- 230000004663 cell proliferation Effects 0.000 description 1
- 238000003570 cell viability assay Methods 0.000 description 1
- 238000012054 celltiter-glo Methods 0.000 description 1
- 230000004640 cellular pathway Effects 0.000 description 1
- 239000001913 cellulose Substances 0.000 description 1
- 229920002678 cellulose Polymers 0.000 description 1
- 238000005119 centrifugation Methods 0.000 description 1
- 201000010881 cervical cancer Diseases 0.000 description 1
- 238000009614 chemical analysis method Methods 0.000 description 1
- 239000007795 chemical reaction product Substances 0.000 description 1
- 230000000973 chemotherapeutic effect Effects 0.000 description 1
- 210000004978 chinese hamster ovary cell Anatomy 0.000 description 1
- 238000004587 chromatography analysis Methods 0.000 description 1
- 238000000576 coating method Methods 0.000 description 1
- 210000001072 colon Anatomy 0.000 description 1
- 208000029742 colonic neoplasm Diseases 0.000 description 1
- 238000004040 coloring Methods 0.000 description 1
- 238000012875 competitive assay Methods 0.000 description 1
- 238000013329 compounding Methods 0.000 description 1
- 238000004590 computer program Methods 0.000 description 1
- 238000012790 confirmation Methods 0.000 description 1
- 238000011109 contamination Methods 0.000 description 1
- 239000000599 controlled substance Substances 0.000 description 1
- 239000006071 cream Substances 0.000 description 1
- 238000004132 cross linking Methods 0.000 description 1
- 150000003983 crown ethers Chemical class 0.000 description 1
- 239000013078 crystal Substances 0.000 description 1
- 210000004748 cultured cell Anatomy 0.000 description 1
- 208000031513 cyst Diseases 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
- UHDGCWIWMRVCDJ-ZAKLUEHWSA-N cytidine Chemical compound O=C1N=C(N)C=CN1[C@H]1[C@H](O)[C@@H](O)[C@H](CO)O1 UHDGCWIWMRVCDJ-ZAKLUEHWSA-N 0.000 description 1
- 230000016396 cytokine production Effects 0.000 description 1
- 230000001461 cytolytic effect Effects 0.000 description 1
- 230000001120 cytoprotective effect Effects 0.000 description 1
- 230000002435 cytoreductive effect Effects 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
- 231100000135 cytotoxicity Toxicity 0.000 description 1
- 238000002784 cytotoxicity assay Methods 0.000 description 1
- 231100000263 cytotoxicity test Toxicity 0.000 description 1
- 239000000412 dendrimer Substances 0.000 description 1
- 229920000736 dendritic polymer Polymers 0.000 description 1
- 238000000432 density-gradient centrifugation Methods 0.000 description 1
- 239000005549 deoxyribonucleoside Substances 0.000 description 1
- 230000001419 dependent effect Effects 0.000 description 1
- ANCLJVISBRWUTR-UHFFFAOYSA-N diaminophosphinic acid Chemical compound NP(N)(O)=O ANCLJVISBRWUTR-UHFFFAOYSA-N 0.000 description 1
- 229960005423 diatrizoate Drugs 0.000 description 1
- 235000014113 dietary fatty acids Nutrition 0.000 description 1
- 238000002050 diffraction method Methods 0.000 description 1
- 238000010790 dilution Methods 0.000 description 1
- 239000012895 dilution Substances 0.000 description 1
- NAGJZTKCGNOGPW-UHFFFAOYSA-K dioxido-sulfanylidene-sulfido-$l^{5}-phosphane Chemical compound [O-]P([O-])([S-])=S NAGJZTKCGNOGPW-UHFFFAOYSA-K 0.000 description 1
- 229940042396 direct acting antivirals thiosemicarbazones Drugs 0.000 description 1
- 231100000676 disease causative agent Toxicity 0.000 description 1
- 230000005750 disease progression Effects 0.000 description 1
- 239000007884 disintegrant Substances 0.000 description 1
- 239000006185 dispersion Substances 0.000 description 1
- PMMYEEVYMWASQN-UHFFFAOYSA-N dl-hydroxyproline Natural products OC1C[NH2+]C(C([O-])=O)C1 PMMYEEVYMWASQN-UHFFFAOYSA-N 0.000 description 1
- 238000012377 drug delivery Methods 0.000 description 1
- 230000009977 dual effect Effects 0.000 description 1
- 229950006432 duligotuzumab Drugs 0.000 description 1
- 239000000975 dye Substances 0.000 description 1
- 230000008030 elimination Effects 0.000 description 1
- 238000003379 elimination reaction Methods 0.000 description 1
- 239000003995 emulsifying agent Substances 0.000 description 1
- 239000000839 emulsion Substances 0.000 description 1
- 230000012202 endocytosis Effects 0.000 description 1
- 210000002919 epithelial cell Anatomy 0.000 description 1
- 210000003743 erythrocyte Anatomy 0.000 description 1
- 201000004101 esophageal cancer Diseases 0.000 description 1
- 210000003238 esophagus Anatomy 0.000 description 1
- 235000019441 ethanol Nutrition 0.000 description 1
- 238000011156 evaluation Methods 0.000 description 1
- 201000005884 exanthem Diseases 0.000 description 1
- 229930195729 fatty acid Natural products 0.000 description 1
- 239000000194 fatty acid Substances 0.000 description 1
- 239000012894 fetal calf serum Substances 0.000 description 1
- 210000002950 fibroblast Anatomy 0.000 description 1
- 239000012997 ficoll-paque Substances 0.000 description 1
- 239000000945 filler Substances 0.000 description 1
- 239000000796 flavoring agent Substances 0.000 description 1
- 235000019634 flavors Nutrition 0.000 description 1
- 239000012530 fluid Substances 0.000 description 1
- MHMNJMPURVTYEJ-UHFFFAOYSA-N fluorescein-5-isothiocyanate Chemical compound O1C(=O)C2=CC(N=C=S)=CC=C2C21C1=CC=C(O)C=C1OC1=CC(O)=CC=C21 MHMNJMPURVTYEJ-UHFFFAOYSA-N 0.000 description 1
- 102000006815 folate receptor Human genes 0.000 description 1
- 108020005243 folate receptor Proteins 0.000 description 1
- 235000003599 food sweetener Nutrition 0.000 description 1
- 229960005102 foscarnet Drugs 0.000 description 1
- 230000005714 functional activity Effects 0.000 description 1
- 229930182830 galactose Natural products 0.000 description 1
- 229910052733 gallium Inorganic materials 0.000 description 1
- 230000005251 gamma ray Effects 0.000 description 1
- 206010017758 gastric cancer Diseases 0.000 description 1
- 229920000159 gelatin Polymers 0.000 description 1
- 235000019322 gelatine Nutrition 0.000 description 1
- 235000011852 gelatine desserts Nutrition 0.000 description 1
- 238000001415 gene therapy Methods 0.000 description 1
- ZDXPYRJPNDTMRX-UHFFFAOYSA-N glutamine Natural products OC(=O)C(N)CCC(N)=O ZDXPYRJPNDTMRX-UHFFFAOYSA-N 0.000 description 1
- 150000002341 glycosylamines Chemical class 0.000 description 1
- 230000012010 growth Effects 0.000 description 1
- 239000003102 growth factor Substances 0.000 description 1
- 238000003306 harvesting Methods 0.000 description 1
- 210000003128 head Anatomy 0.000 description 1
- 201000010536 head and neck cancer Diseases 0.000 description 1
- 208000014829 head and neck neoplasm Diseases 0.000 description 1
- 230000036541 health Effects 0.000 description 1
- 230000003862 health status Effects 0.000 description 1
- 210000003958 hematopoietic stem cell Anatomy 0.000 description 1
- 238000004128 high performance liquid chromatography Methods 0.000 description 1
- 229940088597 hormone Drugs 0.000 description 1
- 239000005556 hormone Substances 0.000 description 1
- 238000001794 hormone therapy Methods 0.000 description 1
- 102000056003 human IL15 Human genes 0.000 description 1
- 239000001257 hydrogen Substances 0.000 description 1
- 125000004435 hydrogen atom Chemical group [H]* 0.000 description 1
- 125000001165 hydrophobic group Chemical group 0.000 description 1
- 229960002591 hydroxyproline Drugs 0.000 description 1
- 239000012216 imaging agent Substances 0.000 description 1
- 230000003053 immunization Effects 0.000 description 1
- 238000002649 immunization Methods 0.000 description 1
- 238000003364 immunohistochemistry Methods 0.000 description 1
- 230000002637 immunotoxin Effects 0.000 description 1
- 229940051026 immunotoxin Drugs 0.000 description 1
- 231100000608 immunotoxin Toxicity 0.000 description 1
- 238000002513 implantation Methods 0.000 description 1
- 230000001976 improved effect Effects 0.000 description 1
- 230000006872 improvement Effects 0.000 description 1
- 230000036512 infertility Effects 0.000 description 1
- 238000003331 infrared imaging Methods 0.000 description 1
- 238000001802 infusion Methods 0.000 description 1
- 230000015788 innate immune response Effects 0.000 description 1
- 229960003786 inosine Drugs 0.000 description 1
- 108040006849 interleukin-2 receptor activity proteins Proteins 0.000 description 1
- 230000004068 intracellular signaling Effects 0.000 description 1
- 238000007917 intracranial administration Methods 0.000 description 1
- 238000007913 intrathecal administration Methods 0.000 description 1
- 238000007914 intraventricular administration Methods 0.000 description 1
- 229960004359 iodixanol Drugs 0.000 description 1
- NBQNWMBBSKPBAY-UHFFFAOYSA-N iodixanol Chemical compound IC=1C(C(=O)NCC(O)CO)=C(I)C(C(=O)NCC(O)CO)=C(I)C=1N(C(=O)C)CC(O)CN(C(C)=O)C1=C(I)C(C(=O)NCC(O)CO)=C(I)C(C(=O)NCC(O)CO)=C1I NBQNWMBBSKPBAY-UHFFFAOYSA-N 0.000 description 1
- 229960001025 iohexol Drugs 0.000 description 1
- NTHXOOBQLCIOLC-UHFFFAOYSA-N iohexol Chemical compound OCC(O)CN(C(=O)C)C1=C(I)C(C(=O)NCC(O)CO)=C(I)C(C(=O)NCC(O)CO)=C1I NTHXOOBQLCIOLC-UHFFFAOYSA-N 0.000 description 1
- 229960004647 iopamidol Drugs 0.000 description 1
- XQZXYNRDCRIARQ-LURJTMIESA-N iopamidol Chemical compound C[C@H](O)C(=O)NC1=C(I)C(C(=O)NC(CO)CO)=C(I)C(C(=O)NC(CO)CO)=C1I XQZXYNRDCRIARQ-LURJTMIESA-N 0.000 description 1
- 229960002603 iopromide Drugs 0.000 description 1
- DGAIEPBNLOQYER-UHFFFAOYSA-N iopromide Chemical compound COCC(=O)NC1=C(I)C(C(=O)NCC(O)CO)=C(I)C(C(=O)N(C)CC(O)CO)=C1I DGAIEPBNLOQYER-UHFFFAOYSA-N 0.000 description 1
- 229960004537 ioversol Drugs 0.000 description 1
- 229940029407 ioxaglate Drugs 0.000 description 1
- TYYBFXNZMFNZJT-UHFFFAOYSA-N ioxaglic acid Chemical compound CNC(=O)C1=C(I)C(N(C)C(C)=O)=C(I)C(C(=O)NCC(=O)NC=2C(=C(C(=O)NCCO)C(I)=C(C(O)=O)C=2I)I)=C1I TYYBFXNZMFNZJT-UHFFFAOYSA-N 0.000 description 1
- 229960002611 ioxilan Drugs 0.000 description 1
- UUMLTINZBQPNGF-UHFFFAOYSA-N ioxilan Chemical compound OCC(O)CN(C(=O)C)C1=C(I)C(C(=O)NCCO)=C(I)C(C(=O)NCC(O)CO)=C1I UUMLTINZBQPNGF-UHFFFAOYSA-N 0.000 description 1
- 229960000310 isoleucine Drugs 0.000 description 1
- AGPKZVBTJJNPAG-UHFFFAOYSA-N isoleucine Natural products CCC(C)C(N)C(O)=O AGPKZVBTJJNPAG-UHFFFAOYSA-N 0.000 description 1
- 235000015110 jellies Nutrition 0.000 description 1
- 210000005067 joint tissue Anatomy 0.000 description 1
- 210000003734 kidney Anatomy 0.000 description 1
- 239000008101 lactose Substances 0.000 description 1
- 229910052746 lanthanum Inorganic materials 0.000 description 1
- 210000000265 leukocyte Anatomy 0.000 description 1
- 150000002632 lipids Chemical class 0.000 description 1
- 201000007270 liver cancer Diseases 0.000 description 1
- 208000014018 liver neoplasm Diseases 0.000 description 1
- 210000004072 lung Anatomy 0.000 description 1
- 201000011649 lymphoblastic lymphoma Diseases 0.000 description 1
- 239000012139 lysis buffer Substances 0.000 description 1
- 239000011777 magnesium Substances 0.000 description 1
- 229910052749 magnesium Inorganic materials 0.000 description 1
- VZCYOOQTPOCHFL-UPHRSURJSA-N maleic acid Chemical compound OC(=O)\C=C/C(O)=O VZCYOOQTPOCHFL-UPHRSURJSA-N 0.000 description 1
- 208000015486 malignant pancreatic neoplasm Diseases 0.000 description 1
- 229910052748 manganese Inorganic materials 0.000 description 1
- 239000011572 manganese Substances 0.000 description 1
- WPBNNNQJVZRUHP-UHFFFAOYSA-L manganese(2+);methyl n-[[2-(methoxycarbonylcarbamothioylamino)phenyl]carbamothioyl]carbamate;n-[2-(sulfidocarbothioylamino)ethyl]carbamodithioate Chemical compound [Mn+2].[S-]C(=S)NCCNC([S-])=S.COC(=O)NC(=S)NC1=CC=CC=C1NC(=S)NC(=O)OC WPBNNNQJVZRUHP-UHFFFAOYSA-L 0.000 description 1
- 230000007246 mechanism Effects 0.000 description 1
- 230000010534 mechanism of action Effects 0.000 description 1
- 201000001441 melanoma Diseases 0.000 description 1
- 238000002844 melting Methods 0.000 description 1
- 230000008018 melting Effects 0.000 description 1
- YACKEPLHDIMKIO-UHFFFAOYSA-N methylphosphonic acid Chemical compound CP(O)(O)=O YACKEPLHDIMKIO-UHFFFAOYSA-N 0.000 description 1
- LSDPWZHWYPCBBB-UHFFFAOYSA-O methylsulfide anion Chemical compound [SH2+]C LSDPWZHWYPCBBB-UHFFFAOYSA-O 0.000 description 1
- 229960004712 metrizoic acid Drugs 0.000 description 1
- GGGDNPWHMNJRFN-UHFFFAOYSA-N metrizoic acid Chemical compound CC(=O)N(C)C1=C(I)C(NC(C)=O)=C(I)C(C(O)=O)=C1I GGGDNPWHMNJRFN-UHFFFAOYSA-N 0.000 description 1
- 108091070501 miRNA Proteins 0.000 description 1
- 239000002679 microRNA Substances 0.000 description 1
- 150000007522 mineralic acids Chemical class 0.000 description 1
- 108091005601 modified peptides Proteins 0.000 description 1
- 239000003068 molecular probe Substances 0.000 description 1
- HPNSFSBZBAHARI-RUDMXATFSA-N mycophenolic acid Chemical compound OC1=C(C\C=C(/C)CCC(O)=O)C(OC)=C(C)C2=C1C(=O)OC2 HPNSFSBZBAHARI-RUDMXATFSA-N 0.000 description 1
- 230000001400 myeloablative effect Effects 0.000 description 1
- 229940031182 nanoparticles iron oxide Drugs 0.000 description 1
- 238000002663 nebulization Methods 0.000 description 1
- 210000003739 neck Anatomy 0.000 description 1
- 239000013642 negative control Substances 0.000 description 1
- 210000000440 neutrophil Anatomy 0.000 description 1
- 229910052759 nickel Inorganic materials 0.000 description 1
- 229910052757 nitrogen Inorganic materials 0.000 description 1
- 229960005419 nitrogen Drugs 0.000 description 1
- 150000003833 nucleoside derivatives Chemical class 0.000 description 1
- 229920002113 octoxynol Polymers 0.000 description 1
- 239000003921 oil Substances 0.000 description 1
- 235000019198 oils Nutrition 0.000 description 1
- 239000002674 ointment Substances 0.000 description 1
- 230000003287 optical effect Effects 0.000 description 1
- 238000005457 optimization Methods 0.000 description 1
- 210000000056 organ Anatomy 0.000 description 1
- 150000007524 organic acids Chemical class 0.000 description 1
- 230000003204 osmotic effect Effects 0.000 description 1
- 230000002611 ovarian Effects 0.000 description 1
- 229910052760 oxygen Inorganic materials 0.000 description 1
- 239000001301 oxygen Substances 0.000 description 1
- QVGXLLKOCUKJST-BJUDXGSMSA-N oxygen-15 atom Chemical compound [15O] QVGXLLKOCUKJST-BJUDXGSMSA-N 0.000 description 1
- 239000003973 paint Substances 0.000 description 1
- 210000000496 pancreas Anatomy 0.000 description 1
- 201000002528 pancreatic cancer Diseases 0.000 description 1
- 208000008443 pancreatic carcinoma Diseases 0.000 description 1
- 238000007911 parenteral administration Methods 0.000 description 1
- 239000006072 paste Substances 0.000 description 1
- 229940049954 penicillin Drugs 0.000 description 1
- 229960003330 pentetic acid Drugs 0.000 description 1
- 229940111202 pepsin Drugs 0.000 description 1
- 229960004624 perflexane Drugs 0.000 description 1
- 210000005259 peripheral blood Anatomy 0.000 description 1
- 239000011886 peripheral blood Substances 0.000 description 1
- 229940124531 pharmaceutical excipient Drugs 0.000 description 1
- 239000000825 pharmaceutical preparation Substances 0.000 description 1
- 230000000144 pharmacologic effect Effects 0.000 description 1
- NBIIXXVUZAFLBC-UHFFFAOYSA-K phosphate Chemical compound [O-]P([O-])([O-])=O NBIIXXVUZAFLBC-UHFFFAOYSA-K 0.000 description 1
- 239000010452 phosphate Substances 0.000 description 1
- PTMHPRAIXMAOOB-UHFFFAOYSA-L phosphoramidate Chemical compound NP([O-])([O-])=O PTMHPRAIXMAOOB-UHFFFAOYSA-L 0.000 description 1
- 230000026731 phosphorylation Effects 0.000 description 1
- 238000006366 phosphorylation reaction Methods 0.000 description 1
- BZQFBWGGLXLEPQ-REOHCLBHSA-N phosphoserine Chemical compound OC(=O)[C@@H](N)COP(O)(O)=O BZQFBWGGLXLEPQ-REOHCLBHSA-N 0.000 description 1
- 239000006187 pill Substances 0.000 description 1
- 210000002381 plasma Anatomy 0.000 description 1
- 210000004180 plasmocyte Anatomy 0.000 description 1
- 229920001481 poly(stearyl methacrylate) Polymers 0.000 description 1
- 238000002264 polyacrylamide gel electrophoresis Methods 0.000 description 1
- 229920000768 polyamine Polymers 0.000 description 1
- 229920000570 polyether Polymers 0.000 description 1
- 229920000656 polylysine Polymers 0.000 description 1
- 238000003752 polymerase chain reaction Methods 0.000 description 1
- 239000000256 polyoxyethylene sorbitan monolaurate Substances 0.000 description 1
- 235000010486 polyoxyethylene sorbitan monolaurate Nutrition 0.000 description 1
- 229920006316 polyvinylpyrrolidine Polymers 0.000 description 1
- 150000004032 porphyrins Chemical class 0.000 description 1
- 238000012636 positron electron tomography Methods 0.000 description 1
- 230000004481 post-translational protein modification Effects 0.000 description 1
- 230000001124 posttranscriptional effect Effects 0.000 description 1
- 239000011591 potassium Substances 0.000 description 1
- 229910052700 potassium Inorganic materials 0.000 description 1
- 230000003389 potentiating effect Effects 0.000 description 1
- 238000004321 preservation Methods 0.000 description 1
- 239000003755 preservative agent Substances 0.000 description 1
- 238000012545 processing Methods 0.000 description 1
- 210000002307 prostate Anatomy 0.000 description 1
- 235000019833 protease Nutrition 0.000 description 1
- 230000004952 protein activity Effects 0.000 description 1
- 108020001580 protein domains Proteins 0.000 description 1
- 229940121649 protein inhibitor Drugs 0.000 description 1
- 239000012268 protein inhibitor Substances 0.000 description 1
- 238000001742 protein purification Methods 0.000 description 1
- PTJWIQPHWPFNBW-GBNDHIKLSA-N pseudouridine Chemical compound O[C@@H]1[C@H](O)[C@@H](CO)O[C@H]1C1=CNC(=O)NC1=O PTJWIQPHWPFNBW-GBNDHIKLSA-N 0.000 description 1
- 239000012521 purified sample Substances 0.000 description 1
- 238000003908 quality control method Methods 0.000 description 1
- 238000001959 radiotherapy Methods 0.000 description 1
- 206010037844 rash Diseases 0.000 description 1
- 239000011541 reaction mixture Substances 0.000 description 1
- 238000010188 recombinant method Methods 0.000 description 1
- 238000011084 recovery Methods 0.000 description 1
- 238000004064 recycling Methods 0.000 description 1
- 239000011347 resin Substances 0.000 description 1
- 229920005989 resin Polymers 0.000 description 1
- 230000004044 response Effects 0.000 description 1
- 108091008146 restriction endonucleases Proteins 0.000 description 1
- 201000006845 reticulosarcoma Diseases 0.000 description 1
- 208000029922 reticulum cell sarcoma Diseases 0.000 description 1
- 108010056030 retronectin Proteins 0.000 description 1
- 239000002342 ribonucleoside Substances 0.000 description 1
- 150000003290 ribose derivatives Chemical group 0.000 description 1
- 210000003705 ribosome Anatomy 0.000 description 1
- 102200120948 rs1057520695 Human genes 0.000 description 1
- 102200120772 rs876661123 Human genes 0.000 description 1
- 239000012266 salt solution Substances 0.000 description 1
- 230000028327 secretion Effects 0.000 description 1
- 238000010187 selection method Methods 0.000 description 1
- 230000035945 sensitivity Effects 0.000 description 1
- 238000012163 sequencing technique Methods 0.000 description 1
- 230000035939 shock Effects 0.000 description 1
- 210000003491 skin Anatomy 0.000 description 1
- 231100000046 skin rash Toxicity 0.000 description 1
- 229910052708 sodium Inorganic materials 0.000 description 1
- 239000011780 sodium chloride Substances 0.000 description 1
- JJGWLCLUQNFDIS-GTSONSFRSA-M sodium;1-[6-[5-[(3as,4s,6ar)-2-oxo-1,3,3a,4,6,6a-hexahydrothieno[3,4-d]imidazol-4-yl]pentanoylamino]hexanoyloxy]-2,5-dioxopyrrolidine-3-sulfonate Chemical compound [Na+].O=C1C(S(=O)(=O)[O-])CC(=O)N1OC(=O)CCCCCNC(=O)CCCC[C@H]1[C@H]2NC(=O)N[C@H]2CS1 JJGWLCLUQNFDIS-GTSONSFRSA-M 0.000 description 1
- 239000007787 solid Substances 0.000 description 1
- 239000007909 solid dosage form Substances 0.000 description 1
- 210000000952 spleen Anatomy 0.000 description 1
- 210000004989 spleen cell Anatomy 0.000 description 1
- 210000003802 sputum Anatomy 0.000 description 1
- 208000024794 sputum Diseases 0.000 description 1
- 206010041823 squamous cell carcinoma Diseases 0.000 description 1
- 230000006641 stabilisation Effects 0.000 description 1
- 238000011105 stabilization Methods 0.000 description 1
- 239000003381 stabilizer Substances 0.000 description 1
- 238000011397 standard salvage chemotherapy Methods 0.000 description 1
- 239000008107 starch Substances 0.000 description 1
- 235000019698 starch Nutrition 0.000 description 1
- 238000007619 statistical method Methods 0.000 description 1
- 210000000130 stem cell Anatomy 0.000 description 1
- 230000004936 stimulating effect Effects 0.000 description 1
- 210000002784 stomach Anatomy 0.000 description 1
- 201000011549 stomach cancer Diseases 0.000 description 1
- 238000003860 storage Methods 0.000 description 1
- 229960005322 streptomycin Drugs 0.000 description 1
- 239000007929 subcutaneous injection Substances 0.000 description 1
- 238000010254 subcutaneous injection Methods 0.000 description 1
- 239000000758 substrate Substances 0.000 description 1
- 239000005720 sucrose Substances 0.000 description 1
- 150000008163 sugars Chemical class 0.000 description 1
- 229910052717 sulfur Inorganic materials 0.000 description 1
- 239000011593 sulfur Substances 0.000 description 1
- 239000000829 suppository Substances 0.000 description 1
- 239000000725 suspension Substances 0.000 description 1
- 238000013268 sustained release Methods 0.000 description 1
- 239000012730 sustained-release form Substances 0.000 description 1
- 239000003765 sweetening agent Substances 0.000 description 1
- 210000001179 synovial fluid Anatomy 0.000 description 1
- 238000003786 synthesis reaction Methods 0.000 description 1
- 230000009885 systemic effect Effects 0.000 description 1
- 101150047061 tag-72 gene Proteins 0.000 description 1
- 229940095064 tartrate Drugs 0.000 description 1
- 230000002381 testicular Effects 0.000 description 1
- 125000005207 tetraalkylammonium group Chemical group 0.000 description 1
- VUYXVWGKCKTUMF-UHFFFAOYSA-N tetratriacontaethylene glycol monomethyl ether Chemical compound COCCOCCOCCOCCOCCOCCOCCOCCOCCOCCOCCOCCOCCOCCOCCOCCOCCOCCOCCOCCOCCOCCOCCOCCOCCOCCOCCOCCOCCOCCOCCOCCOCCOCCO VUYXVWGKCKTUMF-UHFFFAOYSA-N 0.000 description 1
- RYYWUUFWQRZTIU-UHFFFAOYSA-K thiophosphate Chemical compound [O-]P([O-])([O-])=S RYYWUUFWQRZTIU-UHFFFAOYSA-K 0.000 description 1
- ZCUFMDLYAMJYST-UHFFFAOYSA-N thorium dioxide Chemical compound O=[Th]=O ZCUFMDLYAMJYST-UHFFFAOYSA-N 0.000 description 1
- 230000002110 toxicologic effect Effects 0.000 description 1
- 231100000759 toxicological effect Toxicity 0.000 description 1
- 239000003053 toxin Substances 0.000 description 1
- 231100000765 toxin Toxicity 0.000 description 1
- FGMPLJWBKKVCDB-UHFFFAOYSA-N trans-L-hydroxy-proline Natural products ON1CCCC1C(O)=O FGMPLJWBKKVCDB-UHFFFAOYSA-N 0.000 description 1
- VZCYOOQTPOCHFL-UHFFFAOYSA-N trans-butenedioic acid Natural products OC(=O)C=CC(O)=O VZCYOOQTPOCHFL-UHFFFAOYSA-N 0.000 description 1
- 230000009261 transgenic effect Effects 0.000 description 1
- 230000014616 translation Effects 0.000 description 1
- 238000002054 transplantation Methods 0.000 description 1
- QORWJWZARLRLPR-UHFFFAOYSA-H tricalcium bis(phosphate) Chemical compound [Ca+2].[Ca+2].[Ca+2].[O-]P([O-])([O-])=O.[O-]P([O-])([O-])=O QORWJWZARLRLPR-UHFFFAOYSA-H 0.000 description 1
- 102000003298 tumor necrosis factor receptor Human genes 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
- 241001529453 unidentified herpesvirus Species 0.000 description 1
- 241001430294 unidentified retrovirus Species 0.000 description 1
- 229940035893 uracil Drugs 0.000 description 1
- DRTQHJPVMGBUCF-UHFFFAOYSA-N uracil arabinoside Natural products OC1C(O)C(CO)OC1N1C(=O)NC(=O)C=C1 DRTQHJPVMGBUCF-UHFFFAOYSA-N 0.000 description 1
- 229940045145 uridine Drugs 0.000 description 1
- 210000003932 urinary bladder Anatomy 0.000 description 1
- 208000010570 urinary bladder carcinoma Diseases 0.000 description 1
- 210000002700 urine Anatomy 0.000 description 1
- 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 description 1
- 125000002987 valine group Chemical group [H]N([H])C([H])(C(*)=O)C([H])(C([H])([H])[H])C([H])([H])[H] 0.000 description 1
- 210000003462 vein Anatomy 0.000 description 1
- 230000035899 viability Effects 0.000 description 1
- 239000000080 wetting agent Substances 0.000 description 1
- 210000005253 yeast cell Anatomy 0.000 description 1
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/705—Receptors; Cell surface antigens; Cell surface determinants
- C07K14/71—Receptors; Cell surface antigens; Cell surface determinants for growth factors; for growth regulators
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/28—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
- C07K16/2863—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N33/00—Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
- G01N33/48—Biological material, e.g. blood, urine; Haemocytometers
- G01N33/50—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
- G01N33/53—Immunoassay; Biospecific binding assay; Materials therefor
- G01N33/574—Immunoassay; Biospecific binding assay; Materials therefor for cancer
- G01N33/57484—Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/505—Medicinal preparations containing antigens or antibodies comprising antibodies
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/20—Immunoglobulins specific features characterized by taxonomic origin
- C07K2317/24—Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/30—Immunoglobulins specific features characterized by aspects of specificity or valency
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/70—Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/70—Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
- C07K2317/73—Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/70—Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
- C07K2317/73—Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
- C07K2317/732—Antibody-dependent cellular cytotoxicity [ADCC]
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/90—Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
- C07K2317/92—Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2740/00—Reverse transcribing RNA viruses
- C12N2740/00011—Details
- C12N2740/10011—Retroviridae
- C12N2740/16011—Human Immunodeficiency Virus, HIV
- C12N2740/16041—Use of virus, viral particle or viral elements as a vector
- C12N2740/16043—Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N2333/00—Assays involving biological materials from specific organisms or of a specific nature
- G01N2333/435—Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
- G01N2333/475—Assays involving growth factors
- G01N2333/485—Epidermal growth factor [EGF] (urogastrone)
Definitions
- Immune cell products with homogenous expression of tumor targeting chimeric antigen receptors are desirable for clinical evaluation of adoptive therapy strategies to eliminate the product-to-product variability of transgene expression otherwise intrinsic to transduction and other genetic modification procedures without subsequent selection.
- Immunotherapy using genetically redirected immune cells is an attractive approach for treating minimal residual disease in a variety of cancer patients.
- immunologic rejection of cell products expressing antibiotic selection proteins as part of the transduction strategy has impeded this strategy.
- a novel selection marker that is not expressed on human lymphocytes, does not contain endogenous signaling or trafficking function, and is recognized by a known, preferably commercially available, pharmaceutical grade antibody reagent that can be utilized for selection, in vivo tracking, and depletion of transduced cells would be a significant improvement in the art. Disclosed herein, inter alia, are solutions to these and other problems in the art.
- compositions and methods for purification, both in vivo and ex vivo, of genetically modified cells are provided herein.
- the genetically modified cells may be modified by transduction, or any other process that adds, deletes, alters, or disrupts an endogenous nucleotide sequence.
- the genetically modified cells may be transduced T cells with altered activity, including altered immunoactivity.
- a non-immunogenic selection epitope compatible with, for example, immunomagnetic selection, which facilitates immunotherapy in cancer patients without undesirable immunologic rejection of cell products (i.e. as seen when expressing antibiotic selection proteins).
- the non-immunogenic selection epitope is a gene encoding an endogenous cell-surface molecule that is modified or truncated to retain an extracellular epitope recognized by an antibody or functional fragment thereof, and to remove any signaling or trafficking domains and/or any extracellular domains unrecognized by the known antibody.
- the removal of the signaling or trafficking domains and/or any extracellular domains unrecognized by the known antibody renders the endogenous cell- surface molecule inert, which is a desired property for the molecule.
- the non-immunogenic selection epitope may also be used as a selection tool or tracking marker.
- the modified endogenous cell-surface molecule may be, but is not limited to, any cell-surface related receptor, ligand, glycoprotein, cell adhesion molecule, antigen, integrin or cluster of differentiation (CD) that is modified as described herein.
- the modified endogenous cell-surface molecule is a truncated tyrosine kinase receptor.
- the truncated tyrosine kinase receptor is a member of the epidermal growth factor receptor family (e.g., ErbBl, ErbB2, ErbB3, ErbB4).
- Epidermal growth factor receptor also known as EGFR, ErbBl and HER1
- EGFR epidermal growth factor receptor
- HER1 is a cell- surface receptor of the epidermal growth factor family of extracellular ligands. Alterations in EGFR activity have been implicated in certain cancers.
- a gene encoding an EGFR polypeptide is provided that is formed by removal of nucleic acid sequences that encode polypeptides including the membrane distal EGF -binding domain and the cytoplasmic signaling tail (a "truncated EGFR", “tEGFR” or "EGFRt”), but retains the extracellular domain IV epitope recognized by any anti-EGFR antibody (e.g., anti-domain IV EGFR antibody) provided herein including embodiments thereof.
- tEGFR does not include EGFR domain III.
- biotinylated-anti-EGFR antibodies e.g., anti-domain IV EGFR antibodies
- immunomagnetic selection in combination with anti-biotin microbeads successfully will enrich T cells that are lentivirally transduced with EGFRt- containing constructs from as low as 2% of the population to greater than 90% purity without observable toxicity to the cell preparation.
- Constitutive expression of this inert EGFRt molecule does not affect T cell phenotype or effector function as directed by the coordinately expressed chimeric antigen receptor (CAR), CD19R.
- CAR coordinately expressed chimeric antigen receptor
- EGFRt will be shown to have suicide gene potential through anti-EGFR antibodies (e.g., anti-domain IV EGFR antibodies)-mediated antibody dependent cellular cytotoxicity (ADCC) pathways.
- ADCC antibody dependent cellular cytotoxicity
- EGFRt may be used as a non-immunogenic selection tool, tracking marker, and suicide gene for transduced T cells that have immunotherapeutic potential.
- the EGFRt nucleic acid may also be detected by means well known in the art.
- methods of discovering and designing modified, truncated or altered endogenous cell-surface molecules which bind to anti-EGFR antibodies e.g., anti domain IV EGFR antibodies
- the methods include modeling the protein of interest and truncating functional portions, while leaving the antibody -binding portions intact.
- the resulting modified receptor or ligand can be sorted using a labeled antibody and then enriched such that the concentration of the modified receptor or ligand is increased.
- Yet another embodiment provides a method of selecting a transduced T cell including transducing a population of T cells with a modified, truncated or altered endogenous cell-surface molecule gene sequence (e.g., truncated EGFR) and then contacting an antibody that binds the modified ligand or receptor sequence to the transduced T cells.
- a modified receptor sequence is EGFRt
- the antibody is preferably a biotinylated anti- EGFR antibody (e.g., anti-domain IV EGFR antibody).
- the T cells are then sorted by adding anti-biotin microbeads and selected using immunomagnetic separation, adding fluorochrome- conjugated anti-biotin and selecting the T cells using Fluorescence Activated Cell Sorting, or any other reliable method of sorting the cells.
- the modified ligand or receptor sequences such as the EGFRt sequence, may be contained in a suitable transfer vehicle such as a lentiviral vector.
- FIG. 1. is a schematic representation of the EGFR protein domains. Mutations within the extracellular domains (ECD) are shown, several of which are shown to induce EGFR auto-phosphorylation, which in turn promotes cellular transformation.
- FIG. 2. shows the superposition of structures of therapeutic anti-EGFR antibodies in complex with domain III of EGFR. The binding location of the antibodies is consistent with inhibition of EGF binding to EGFR.
- the anti-EGFR antibody Fabs are shown as ribbon diagrams and EGFR is shown as a surface rendering.
- the PDB accession codes for the overlaid structures are as labeled.
- FIG. 3. shows non-antibody EGFR-binding therapeutics in complex with EGFR domain III.
- the non-antibody EGFR-binding therapeutics are depicted as ribbon diagrams and EGFR is shown as a surface rendering.
- the PDB accession codes for the structures are as labeled.
- FIG. 4. shows the superposition of various Her2 targeting biologies with Her2.
- the binding of trastuzumab to the juxtamembrane domain (domain IV) of Her2 is labeled.
- Her2 is represented as a surface rendering and Her2 targeting biologies are shown as ribbon diagrams.
- FIG.s 5A-5C show the correlation between T cell activation and the proximity of the epitope and cell membrane.
- FIG. 5A illustrates the distance between the cancer cell membrane and T cell membrane upon formation of the T cell receptor (TCR)/CD3 complex involved in antibody-dependent cellular cytotoxicity (ADCC) activity.
- FIG. 5B demonstrates that the geometry of the Her2 antigen in respect to the cancer cell membrane is critical to achieve effective ADCC activity. Targeting of domains closer to the cancer cell membrane results in greater ADCC effect while targeting the domain furthest from the membrane does not result in effective ADCC activity.
- FIG. 5C. is a graph illustrating the results from a T cell activation assay, showing that targeting of antigen domains closer to the cell membrane effectively produces high ADCC activity compared to antigen domains further from the membrane.
- FIG. 6. are schematics showing the constructs of chimeric human EGFR domain IV proteins D4-IgG2a, EGFR-D4-IgG2a and EGFR-IGA1.
- FIG. 7. are chromatograms depicting purification of mouse D4-IgG2A (top panel) and human EGFR-D4-IgAl (bottom panel) with size exclusion chromatography. The chromatography indicates the antibodies were purified to substantial homogeneity.
- FIG. 8. illustrates results from an ELISA experiment identifying six hybridoma clones that bind human EGFR-IgA protein.
- EGFR-IgA proteins were immobilized in microwells and were subsequently incubated with hybridoma culture supernatant. Results indicate that the 5C8 clone had the highest antibody titer.
- FIG. 9. shows results from a flow cytometry experiment testing binding of hybridoma clones to SKOV3 cells. Results indicate that the 5C8 antibody clone binds to the ovarian cancer cell line SKOV3 compared to an isotype control.
- FIG. 10 shows results from an ELISA experiment illustrating the specificity of the 5C8 clone for binding with various recombinant proteins. The results indicate that the clone is specific for binding to D4-IgA protein which includes EGFR domain IV fused with human IgAl.Fc, and EGFR-His.
- FIG. 11. illustrates results from a flow cytometry experiment showing that the 5C8 clone is specific for EGFR-expressing cells, for example the breast cancer cell line SKBR3. As expected, the 5C8 antibody does not bind to Jurkat cells which do not express EGFR.
- FIG. 12. illustrates results from an ELISA experiment showing that the Ig isotypes of the 5C8 clone are g ⁇ and l.
- FIG.s 13A and 13B show purification and characterization of the anti-domain IV EGFR antibody clone EGFRD4-5C8.
- FIG. 13A is a chromatogram depicting purification of EGFRD4-5C8 using size exclusion chromatography
- FIG. 13B. shows a reducing (R) and non-reducing (NR) gel characterizing the purified product. As expected, the non-reducing gel showed a product at approximately 150 kDa while the reducing gel showed two bands at approximately 25 kDa and 50 kDa.
- FIG.s 14A-14C. are sensorgrams from SPR experiments that depict binding of FIG. 14A. EGFR-D4-5C8 antibody, FIG. 14B. cetuximab Fab, and FIG. 14C. wild type traszumab to EGFR domain IV. EGFR-D4-5C8 was shown to be captured by immobilized EGFR domain IV, while cetuximab Fab and wild type trastuzumab do not show substantial binding to domain IV of EGFR.
- FIG.s 15A-15C. are sensorgrams from SPR experiments showing binding of FIG. 15A. EGFR-D4-5C8 Fab, FIG. 15B. cetuximab Fab, and FIG. 15C. wild type trastuzumab Fab to EGFR domain IV. As expected, EGFR-D4-5C8 Fab was shown to be captured by immobilized EGFR domain IV, while cetuximab Fab and wild type trastuzumab Fab do not show substantial binding to domain IV of EGFR. [0027] FIG. 16.
- FIGS. 1-10 are representative images of Western blots showing detection of phosphorylated-EGFR (phsopho-EGFR), phosphorylated Akt (phospho-Akt), or b-actin (positive control) upon incubating the human ovarian carcinoma cell line OVCAR3 with no antibody, EGF, cetuximab or the EGFR-D4-5C8 antibody clone.
- cetuximab no phosphor-EGFR was detected, as expected.
- phospho-EGFR was detected upon addition of EGFR-D4-5C8.
- the results indicate that cetuximab and EGFR- D4-5C8 affect different cellular pathways.
- FIG. 17 shows flow cytometry data detecting the binding of cetuximab (top row) or EGFR-D4-5C8 (bottom row) to MDA-MB-468, SCOV3 or OVCAR3 cells.
- the similar binding abilities of cetuximab and EGFR-D4-5C8 to the cells may be attributed to the fluorophore intensity and binding affinity of the secondary antibody PE anti-Fc.
- FIG. 18A show results from an ADCC assay.
- MDA-MB-468 cells and SKOV3 cells were incubated with Jurkat cells expressing NFAT-regulated luciferase and either 5C8 or cetuximab antibodies at different concentrations. Luciferase substrate was added to each solution, and ADCC activity was measured based on luminescence intensity.
- FIG. 18B is flow cytometry data showing that cetuximab, anti-HER3 antibody and a dual anti-EGFR/HER3 meditope-enabled antibody bind to the bladder carcinoma cell line 486 and the ovarian cancer cell line SKOV3 (left panel); the EGFR-D4-5C8 antibody also shows binding ability to both cell lines (right panel).
- FIG.s 19A-19G. are results from ADCC assays conducted with Jurkat cells and various EGFR-expressing cancer cell lines incubated with the 5C8 antibody clone or cetuximab.
- FIG. 19A MDA-MB-468 cells
- FIG. 19B SW48 cells
- FIG. 19C SKOV3 cells
- FIG. 19D A549 cells
- FIG. 19E HCT116 cells were incubated with 5C8 or cetuximab antibody at different concentrations.
- luciferase substrate was added in each well to react with luciferase expressed by Jurkat cells.
- ADCC activity was measured based on luminescence intensity. Results from the assays show that the 5C8 antibody exerts stronger ADCC effects than cetuximab.
- FIG. 19F. shows the combined ADCC for the 5C8 clone
- FIG. 19G shows the combined ADCC for cetuximab.
- FIG. 20 shows binding of EGFR-targeting antibodies to EGFR-expressing cancer cell lines, as determined by flow cytometry.
- Cells were incubated with 10 ug/ml 5C8 or 10 ug/ml cetuximab for 30 minutes, and subsequently washed, followed by staining of cells with anti-kappa-Alexa-647 secondary antibody. Histogram analysis (top panel) and median fluorescence intensity of each cell line before and after antibody binding to the cells (bottom panel) is depicted.
- FIG. 21 is the time course for in vivo ADCC experiments using animal xenograft models showing. EGFR + tumor killing.
- FIG. 22 illustrates results from an ADCC experiment using 5C8-IgGl, 5C8-IgG2a, or cetuximab-IgG2a antibodies. Results indicate the 5C8-IgG2a antibody can eradicate tumors in vivo, while the 5C8-IgGl antibody does not have a therapeutic effect as compared with the vehicle control.
- FIG. 23 is a phylogram showing sequence distance relationships between the identified anti-domain IV EGFR antibody clones.
- FIG. 24 Depicted are phylograms comparing the sequence relationships between the identified anti-domain IV EGFR antibody clones and the sequence alignment of various antibody clones.
- FIG.s 25A-25F. are chromatograms showing purification of anti-domain IV EGFR antibody clones FIG. 25A. EGFRD4-7Ab, FIG. 25B. EGFRD4-28Ab, FIG. 25C. EGFRD4- 30Ab, FIG. 25D. EGFRD4-31Ab, and FIG. 25E. EGFRD4-34Ab by size exclusion chromatography; and FIG. 25F. a representative image of a non-reduced and reduced gel showing separation of the purified products. As expected, a higher molecular weight band for each sample is shown on the non-reducing gel, while the reducing gel shows two bands at approximately 50 kDa and 25 kDa for each antibody sample.
- FIG.s 26A-26F show sensorgrams from SPR experiments depicting binding of various anti-domain IV EGFR antibodies to domain IV of EGFR.
- EGFR domain IV was immobilized on an CM5 chip at a density of 500 RU through EDC/NHS coupling.
- the EGFRD4 (IgG) antibodies FIG. 26A. EGFRD4-7Ab, FIG. 26B. EGFRD4-28Ab, FIG. 26C. EGFRD4-30Ab, FIG. 26D. EGFRD4-31Ab, FIG. 26E. EGFRD4-34Ab, and FIG. 26F.
- EGFRD4-5C8 were prepared in HBS-EP + running buffer and were injected at concentrations of 30 nM, 10 nM, and 3 nM at 25 °C.
- FIG. 27. are sensorgrams from SPR experiments showing binding of the EGFRD4- 5C8 clone to EGFR domain IV.
- EGFR domain IV was immobilized on an CM5 chip at a density of 500 RU through EDC/NHS coupling, and various concentrations of EGFRD4-5C8 were prepared in HBS-EP + running buffer and injected.
- SPR binding curves for 30 nM, 10 nM, 3 nM and 1 nM EGFRD4-5C8 are shown (top panel), in addition to a magnified portion of the sensorgram showing binding curves for 10 nM, 3 nM and 1 nM EGFRD4-5C8 (bottom panel).
- FIGS. 28A illustrates the selection of EGFRt.sup.+ T cells using biotinylated an anti-EGFR antibody (e.g., anti-domain IV EGFR antibody) as provided herein.
- an anti-EGFR antibody e.g., anti-domain IV EGFR antibody
- a schematic is shown of the EGFR antibody biotinylation and reformulation process.
- FIG. 28B depicts schematics of both the immunomagnetic (top) and the fluorescence activated cell sorting (bottom) EGFRt selection procedures.
- FIG. 28C depicts schematics of the CD19CAR-T2A-EGFRt (left) and CD19CAR- T2A-EGFRt-IMPDH2dm (right) constructs contained in lentiviral vectors used for the immunomagnetic selection of various T cell lines lentivirally transduced with CAR and EGFRt containing constructs. Codon optimized sequence portions of the CD19-specific, CD28 co-stimulatory CAR, followed by the self-cleavable T2A, EGFRt and IMPDH2dm selection markers are indicated, along with the Elongation Factor 1 promoter sequences (EF- lp), and the GCSFR alpha chain signal sequences (GCSFRss, which directs surface expression).
- EF- lp Elongation Factor 1 promoter sequences
- GCSFRss GCSFR alpha chain signal sequences
- FIG. 29 is the nucleotide (sense strand is SEQ ID NO:277, antisense strand is SEQ ID NO:278) sequence and amino acid (SEQ ID NO:279) sequences of GMCSFR alpha chain signal sequence linked to EGFRt including domain III and domain IV.
- the GMCSFR alpha chain signal sequence, which directs surface expression, is encoded by nucleotides 1-66.
- EGFRt including domain III and IV is encoded by nucleotides 67-1071.
- FIG. 30 is the nucleotide (sense strand is SEQ ID NO: 280, antisense strand is SEQ ID NO:281) sequence and amino acid (SEQ ID NO:282) sequences of CD19R-CD28gg- Zeta(CO)-T2A-EGFRt.
- CD19R-CD28gg-Zeta(CO) is encoded by nucleotides 1-2040;
- T2A is encoded by nucleotides 2041-2112;
- GMCSFR is encoded by nucleotides 2113-2178;
- EGFRt including domain III and domain IV is encoded by nucleotides 2179-3186.
- FIG.s 31A and 31B illustrate FIG. 31A. data showing thermal stability curves of EGFR D4 5C8Fab, EGFR D428Fab, EGFR D430Fab, EGFR D4 31Fab, EGFR D4 34 Fab and meTras 183E Fab (meditope-enabled trastuzumab), and FIG. 31B. the melting point (T m ) of each Fab domain.
- T m melting point
- FIG. 32 shows binding of EGFR-targeting antibodies to EGFR-expressing cancer cell lines MDA-MB-4 ⁇ 3 ⁇ 4 SKOV3 and OVCAR-3, as determined by flow cytometry. Because the 5C8 antibody has lower binding affinity, more antibody is needed to detect a shift in intensity.
- FIG.s 33A-33C shows binding of EGFR D4 targeting antibodies and cetuximab to FIG. 33 A.
- FIG. 33B SKOV3, OVCAR-3
- FIG. 33C U87, and U87-EGFRviii cells, as determined by flow cytometry. The cells were detached by trypsin and incubated with 1, 10, or 100 ug/ml EGFR Ab in 2% BSA in PBS at 4 °C. After 30 minutes, cells were washed with 2% BSA in PBS.
- the cells were then stained with secondary antibodies, either anti-Fc PE for MDA-MB-468, MDA-MB-231, SKOV3 and OVCAR3 cells, or anti-Fc Alexa-647 for U87 and U87-EGFRviii cells at 4 °C for 30 min.
- the cells were analyzed by flow cytometry after washing.
- FIG.s 34A-34D illustrates results from cell viability assays conducted with cetuximab, EGFR-D4 targeting antibody, or a combination thereof and cancer cell lines
- Cells were incubated with different concentrations of the indicated Ab for 72h and cell viability was assessed using the Promega CellTiter-Glo® Luminescence kit based on the manufacture’s instructions.
- Cetuximab was selected to intentionally block EGF from binding to EGFR by Mendelsohn (PMID: 6298788) and Rodeck (PMID: 2250044). The reasoning was that cancer cells overexpressing EGFR (e.g.
- FIG. 34D is a schematic showing a mechanism of resistance to domain III targeting therapeutics.
- CTX selection which prevents EGF binding by interacting with domain III of tethered EGFR, can lead to cells aquiring EGFR mutations (G33S, N56K) in domain I, which leads to ligand-independent activation and prevents receptor intemalization/degradation. Mutant EGFR does not bind CTX since it is ‘trapped’ in the open confirmation, leading to CTX resistance (PLoS One. 2020 Feb 18;15(2):e0229077).
- FIG.s 35A and 35B illustrate results from ADCC assays with Jurkat cells expressing immunologulin receptor CD16-158F and FIG. 35 A. MDA-MB-468 cells and FIG. 35B. SKOV3 cells.
- ADCC predominantly works through the FcRIIIa receptor expressed on NK cells. Tumor cells (2.5e4) were seeded in a 96-well white wall plate on day 1 and allowed to attach overnight. On day 2, medium was removed from the plate and Jurkat cells (le5) expressing CD16-158F were added in each well with Ab at the indicated concentration. The final volume was 60ul per well. After incubation for 6h, 50ul of luciferase substrate was added in each well and luminescence was measured immediately.
- FIG.s 36A-36E illustrates binding of EGFR-targeting antibodies to EGFR expressing cancer cell lines and ADCC activity through Jurkat cells expressing immunoglobulin receptor CD16-158V.
- FIG. 36A shows results from FACS studies assessing binding of EGFR-targeting antibodies to cancer cells
- FIG. 36B shows MFI indicative of antibody binding.
- Cells were detached by trypsin and incubated with 10 ug/ml cetuximab or 5C8 in 2% BSA in PBS at 4 °C. After 30 min, cells were washed by 2% BSA in PBS followed by staining cells with anti-Fc Alexa-647 secondary antibody at 4 °C for 30 min.
- FIG. 36C EGFR D4 5C8 antibody
- FIG. 36D Cetuximab.
- Tumor cells 2.5e4 were seeded in a 96 well white wall plate on day 1 and allowed to attach overnight. On day 2, medium was removed from the plate and Jurkat cells (le5) expressing CD 16- 158V were added to each well with Ab at the indicated concentration. The final volume was 60ul per well. After incubation for 6h, 50ul of luciferase substrate was added in each well and luminescence was measured immediately.
- FIG.s 32A and 32B CD16 polymorphism can alter ADCC activity.
- FIG. 36E illustrates FACS data showing CD16-158V expression on Jurkat cells used in this study.
- FIG.s 37A-37E shows results of ADCC assays through Jurkat cells expressing CD 16- 158V and FIG. 37A. MDA-MB-468, FIG. 37B. SCOV3, FIG. 37C. SW48, FIG. 37D. A549 and FIG. 37E. HCT-116 cells.
- the data show a direct comparison of ADCC using EGFR D4-targeting antibody and Cetuximab, and show that the EGFR D4 targeting antibody is significantly more effective than Cetuximab.
- Tumor cells (2.5e4) were seeded in 96 well white wall plate on day 1 and allowed to attach overnight.
- FIG.s 38A-38C illustrate EGFR binding to triple-negative breat cancer (TNBC) cell lines and ADCC activity.
- FIG. 38A shows FACS data assessing various concenrations of indicated antibody binding to MDA-MB-468 and MDA-MB-231 cells. Cells were detached by trypsin and incubated with 1, 10, or 100 ug/ml EGFR Ab in 2% BSA in PBS at 4 °C. After 30 min, the cells were washed by 2% BSA in PBS, followed by staining with anti- Fc PE secondary antibody at 4 °C for 30 min. Cells were analyzed by flow cytometry after after washing.
- FIG. 38B shows FACS data assessing various concenrations of indicated antibody binding to MDA-MB-468 and MDA-MB-231 cells. Cells were detached by trypsin and incubated with 1, 10, or 100 ug/ml EGFR Ab in 2% BSA in PBS at 4 °C. After 30 min, the cells were
- FIG. 38C illustrates ADCC activity with Jurkat cells expressing high affinity CD16-158V.
- ADCC assays were performed by co-culture of CD 16-expressing Jurkat cells (le5) and EGFR-expressing cancer cells (2.5e4) in the presence EGFR Ab at the indiated concentrations. The final incubation volume was 60ul. After 6h incubation, 50ul luciferase substrate was added to each well to react with luciferase expressed by the Jurkat cells. ADCC activity was measured based on luminescence intensity. The results show that CD 16- 158V overexpressed in Jurkat cells had higher affinity to Fc compared to CD16-158F, and therefore induced higher ADCC activity.
- FIG.s 39A-39C show ADCC activity through high affinity and low affinity CD 16 receptors.
- FIG. 39A illustrates binding of Cetuximab and EGFR D4 targeting antibodies to MDA-MB-468 cells. Cells were detached by trypsin and incubated with 1, 10, or 100 ug/ml EGFR Ab in 2% BSA in PBS at 4 °C. After 30 min, cells were washed by 2% BSA in PBS followed by staining cells with anti-Fc PE secondary antibody at 4 °C for 30 min. Cells were analyzed by flow cytometry after washing.
- FIG. 39B shows results from ADCC assays through high affinity CD 16-158V and FIG. 39C.
- ADCC assays were performed by co-culture of CD 16-expressing Jurkat cells (le5) and EGFR-expressing cancer cells (2.5e4) in the presence of EGFR- targeting Ab at different concentrations. The final incubation volume is 60ul. After 6h incubation, 50ul luciferase substrate was added in each well to react with luciferase expressed by Jurkat cells. ADCC activity was measured based on luminescence intensity. The results showed that CD16-158V overexpressed in Jurkat cells had higher affinity to Fc compared to CD16-158F, and therefore induced higher ADCC activity.
- FIG. 40 illustrates ADCC results through PBMC.
- PBMC peripheral blood mononuclear cell
- MDA-MB-468 tumor cells le4
- E:T ratio 20:1.
- the left panel illustrates the percentage of tumor cells remaining after PBMC treatment, with control tumor cells (no treatment) set to 100%.
- the right panel illustrates the percentage of tumor cells killed by PBMC.
- PBMC #1, PBMC #2 and PBMC #3 were from different donors.
- the previous ADCC assays were based on engineered Jurkat cells that quantified ADCC activation through NFAT driven luciferase expression. This method is consistent and efficient.
- PBMC isolated from patients can be used, and cell killing measured.
- the left panel shows the number of viable tumor cells after treatment with PBMC.
- cetuximab kills a fraction of the cells. Without wishing to be bound by theory, this may be due to EGF blockade and onco-addiction.
- the presence of the Ab further reduces the number of viable cells, although it is noted that the PBMC alone reduce the number of tumor cells.
- the right panel represents the same results, but is represented as percent cell death.
- FIG.s 41A-41C illustrates results from ADCC assays through PBMC with cell lines FIG. 41A. MDA-MB-468, FIG. 41B. U87 and FIG. 41C. U87-EGFRviii.
- PBMC peripheral blood mononuclear cells
- FIG. 41B PBMC
- FIG. 41C U87-EGFRviii.
- PBMC peripheral blood mononuclear cell
- Tumor cells without any treatment were set as 100%.
- the E:T ratio was 20:1.
- the results illustrate that Cetuximab barely affected cell viability in the absence of PBMCs.
- the addition of only PBMC dramatically reduced cell viability for all cell lines studies.
- Results using the U87 line are compelling - incubation with only PBMC drastically reduces viability.
- Without PBMC addition of the 5C8 antibody and Cetuximab do not result in significant decreases in cell viability. Additional studies are performed with these and other cell lines to assess conditions including receptor environment, recycling, and other intrinsic
- FIG.s 42A-42D show percent cell killing by PBMC as assessed by ADCC assys for FIG. 42A. MDA-MB-468, FIG. 42B. U87 and FIG. 42C. U87-EGFRviii cells, and FIG. 42D. FACS data showing binding of Cetuximab and EGFR D45C8 to the cell lines MDA- MB-468, U87 and U87-EGFRviii.
- PBMC 2e5
- tumor cells le4 were incubated for 6h with or without indicated Ab. Tumor cells without any treatment were set as 100%.
- the E:T ratio was 20:1.
- cells were detached by trypsin and incubated with 1, 10, or 100 ug/ml EGFR Ab in 2% BSA in PBS at 4 °C. After 30 min, cells were washed by 2% BSA in PBS followed by staining cells with anti-Fc PE (for MDA-MB-468) or anti-Fc Alexa-647 (for U87 and U87-EGFRviii) secondary antibodies at 4 °C for 30 min. Cells were analyzed by flow cytometry after washing.
- FIG. 43 shows a treatment plan for animal xenograft models for EGFR + tumor killing to assess in vivo ADCC.
- mouse strains SCID BALB/c- IglV’ scld ) were used.
- mice were divided into the following treatment groups: PBS, 5 mg/kg, 5 mice; 5C8-IgGl, 5 mg/kg, 5 mice; 5C8-IgG2a, 5 mg/kg, 5 mice; Cetuximab-IgG2a, 5 mg/kg, 5 mice.
- the Ab were delivered intraperitoneally.
- the weight of each mouse was ⁇ 20 mg, and antibody was administered at -100 mg/mouse.
- FIG. 44 shows the MDA-MB-468 xenograft-tumor volume of mice receiving various antibody treatments or PBS vehicle control, as indicated.
- the treatment groups were: PBS, 5 mg/kg, 5 mice; 5C8-IgGl, 5 mg/kg, 5 mice; 5C8-IgG2a, 5 mg/kg, 5 mice; Cetuximab-IgG2a, 5 mg/kg, 5 mice.
- the Ab were delivered intrapen toneallv.
- the weight of each mouse was ⁇ 20 mg, and antibody was administered at -100 mg/mouse.
- FIG. 45 shows a treatment plan for animal xenograft models for EGFR + tumor killing to assess in vivo ADCC.
- mouse strains SCID BALB/c-Igh* scld ) were used.
- mice were divided into the following treatment groups: PBS, 5 mg/kg, 4 mice; 5C8-IgGl, 5 mg/kg, 4 mice; 5C8-IgG2a, 5 mg/kg, 4 mice.
- the antibodies were delivered intraperitoneally. The weight of each mouse was -20 mg, and antibody was administered at -100 mg/mouse.
- FIG. 46 shows the MDA-MB-468 xenograft-tumor volume of mice receiving various antibody treatments or PBS vehicle control, as indicated. It was noted that the group treated with Cetuximab-IgG2a had abdominal distension. This was possibly due to endotoxin contamination for Cetuximab IgG2a Ab. Thus, the mouse group treated with cetuximab- IgG2a was removed from this data set. It was noted that endotoxin units for Cetuximab IgG2a was 0.112 EU/ug, while endotoxin units for 5C8 IgGl was 0.016 EU/ug. The tumors were allowed to grow to a large size before initiating the treatment. A murine version of cetuximab was used for this study. However, subsequent testing of the protein sample indicated substantial amounts of endotoxin, as noted above. Replicates of this study are underway.
- Nucleic acid refers to nucleotides (e.g., deoxyribonucleotides or ribonucleotides) and polymers thereof in either single-, double- or multiple-stranded form, or complements thereof; or nucleosides (e.g., deoxyribonucleosides or ribonucleosides). In embodiments, “nucleic acid” does not include nucleosides.
- polynucleotide oligonucleotide,” “oligo” or the like refer, in the usual and customary sense, to a linear sequence of nucleotides.
- nucleoside refers, in the usual and customary sense, to a glycosylamine including a nucleobase and a five-carbon sugar (ribose or deoxyribose).
- nucleosides include, cytidine, uridine, adenosine, guanosine, thymidine and inosine.
- nucleotide refers, in the usual and customary sense, to a single unit of a polynucleotide, i.e., a monomer. Nucleotides can be ribonucleotides, deoxyribonucleotides, or modified versions thereof.
- polynucleotides contemplated herein include single and double stranded DNA, single and double stranded RNA, and hybrid molecules having mixtures of single and double stranded DNA and RNA.
- nucleic acid e.g. polynucleotides contemplated herein include any types of RNA, e.g. mRNA, siRNA, miRNA, and guide RNA and any types of DNA, genomic DNA, plasmid DNA, and minicircle DNA, and any fragments thereof.
- duplex in the context of polynucleotides refers, in the usual and customary sense, to double strandedness. Nucleic acids can be linear or branched.
- nucleic acids can be a linear chain of nucleotides or the nucleic acids can be branched, e.g., such that the nucleic acids comprise one or more arms or branches of nucleotides.
- the branched nucleic acids are repetitively branched to form higher ordered structures such as dendrimers and the like.
- Nucleic acids can include one or more reactive moieties.
- the term reactive moiety includes any group capable of reacting with another molecule, e.g., a nucleic acid or polypeptide through covalent, non-covalent or other interactions.
- the nucleic acid can include an amino acid reactive moiety that reacts with an amio acid on a protein or polypeptide through a covalent, non-covalent or other interaction.
- the terms also encompass nucleic acids containing known nucleotide analogs or modified backbone residues or linkages, which are synthetic, naturally occurring, and non- naturally occurring, which have similar binding properties as the reference nucleic acid, and which are metabolized in a manner similar to the reference nucleotides.
- Examples of such analogs include, without limitation, phosphodiester derivatives including, e.g., phosphoramidate, phosphorodiamidate, phosphorothioate (also known as phosphothioate having double bonded sulfur replacing oxygen in the phosphate), phosphorodithioate, phosphonocarboxylic acids, phosphonocarboxylates, phosphonoacetic acid, phosphonoformic acid, methyl phosphonate, boron phosphonate, or O-methylphosphoroamidite linkages (see Eckstein, OLIGONUCLEOTIDES AND ANALOGUES: A PRACTICAL APPROACH, Oxford University Press) as well as modifications to the nucleotide bases such as in 5-methyl cytidine or pseudouridine.; and peptide nucleic acid backbones and linkages.
- phosphodiester derivatives including, e.g., phosphoramidate, phosphorodiamidate, phosphorothioate (also known as phosphothio
- nucleic acids include those with positive backbones; non-ionic backbones, modified sugars, and non-ribose backbones (e.g. phosphorodiamidate morpholino oligos or locked nucleic acids (LNA) as known in the art), including those described in U.S. Patent Nos. 5,235,033 and 5,034,506, and Chapters 6 and 7, ASC Symposium Series 580, CARBOHYDRATE MODIFICATIONS IN ANTISENSE RESEARCH, Sanghui & Cook, eds. Nucleic acids containing one or more carbocyclic sugars are also included within one definition of nucleic acids.
- LNA locked nucleic acids
- Modifications of the ribose-phosphate backbone may be done for a variety of reasons, e.g., to increase the stability and half-life of such molecules in physiological environments or as probes on a biochip.
- Mixtures of naturally occurring nucleic acids and analogs can be made; alternatively, mixtures of different nucleic acid analogs, and mixtures of naturally occurring nucleic acids and analogs may be made.
- the intemucleotide linkages in DNA are phosphodiester, phosphodiester derivatives, or a combination of both.
- Nucleic acids can include nonspecific sequences.
- nonspecific sequence refers to a nucleic acid sequence that contains a series of residues that are not designed to be complementary to or are only partially complementary to any other nucleic acid sequence.
- a nonspecific nucleic acid sequence is a sequence of nucleic acid residues that does not function as an inhibitory nucleic acid when contacted with a cell or organism.
- a polynucleotide is typically composed of a specific sequence of four nucleotide bases: adenine (A); cytosine (C); guanine (G); and thymine (T) (uracil (U) for thymine (T) when the polynucleotide is RNA).
- A adenine
- C cytosine
- G guanine
- T thymine
- U uracil
- T thymine
- polynucleotide sequence is the alphabetical representation of a polynucleotide molecule; alternatively, the term may be applied to the polynucleotide molecule itself. This alphabetical representation can be input into databases in a computer having a central processing unit and used for bioinformatics applications such as functional genomics and homology searching.
- Polynucleotides may optionally include one or more non-standard nucleotide(s), nucleotide analog(s) and/or modified nucleo
- complement refers to a nucleotide (e.g., RNA or DNA) or a sequence of nucleotides capable of base pairing with a complementary nucleotide or sequence of nucleotides.
- a complement may include a sequence of nucleotides that base pair with corresponding complementary nucleotides of a second nucleic acid sequence.
- the nucleotides of a complement may partially or completely match the nucleotides of the second nucleic acid sequence. Where the nucleotides of the complement completely match each nucleotide of the second nucleic acid sequence, the complement forms base pairs with each nucleotide of the second nucleic acid sequence. Where the nucleotides of the complement partially match the nucleotides of the second nucleic acid sequence only some of the nucleotides of the complement form base pairs with nucleotides of the second nucleic acid sequence.
- Examples of complementary sequences include coding and a non-coding sequences, wherein the non-coding sequence contains complementary nucleotides to the coding sequence and thus forms the complement of the coding sequence.
- a further example of complementary sequences are sense and antisense sequences, wherein the sense sequence contains complementary nucleotides to the antisense sequence and thus forms the complement of the antisense sequence.
- sequences may be partial, in which only some of the nucleic acids match according to base pairing, or complete, where all the nucleic acids match according to base pairing.
- two sequences that are complementary to each other may have a specified percentage of nucleotides that are the same (i.e., about 60% identity, preferably 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or higher identity over a specified region).
- amino acid refers to naturally occurring and synthetic amino acids, as well as amino acid analogs and amino acid mimetics that function in a manner similar to the naturally occurring amino acids.
- Naturally occurring amino acids are those encoded by the genetic code, as well as those amino acids that are later modified, e.g., hydroxyproline, g- carboxyglutamate, and O-phosphoserine.
- Amino acid analogs refers to compounds that have the same basic chemical structure as a naturally occurring amino acid, i.e., an a carbon that is bound to a hydrogen, a carboxyl group, an amino group, and an R group, e.g., homoserine, norleucine, methionine sulfoxide, methionine methyl sulfonium. Such analogs have modified R groups (e.g., norleucine) or modified peptide backbones, but retain the same basic chemical structure as a naturally occurring amino acid.
- Amino acid mimetics refers to chemical compounds that have a structure that is different from the general chemical structure of an amino acid, but that functions in a manner similar to a naturally occurring amino acid.
- the terms “non-naturally occurring amino acid” and “unnatural amino acid” refer to amino acid analogs, synthetic amino acids, and amino acid mimetics which are not found in nature.
- Amino acids may be referred to herein by either their commonly known three letter symbols or by the one-letter symbols recommended by the IUPAC-IUB Biochemical Nomenclature Commission. Nucleotides, likewise, may be referred to by their commonly accepted single-letter codes.
- polypeptide refers to a polymer of amino acid residues, wherein the polymer may In embodiments be conjugated to a moiety that does not consist of amino acids.
- the terms apply to amino acid polymers in which one or more amino acid residue is an artificial chemical mimetic of a corresponding naturally occurring amino acid, as well as to naturally occurring amino acid polymers and non-naturally occurring amino acid polymers.
- a “fusion protein” refers to a chimeric protein encoding two or more separate protein sequences that are recombinantly expressed as a single moiety.
- amino acid or nucleotide base "position" is denoted by a number that sequentially identifies each amino acid (or nucleotide base) in the reference sequence based on its position relative to the N-terminus (or 5'-end). Due to deletions, insertions, truncations, fusions, and the like that must be taken into account when determining an optimal alignment, in general the amino acid residue number in a test sequence determined by simply counting from the N-terminus will not necessarily be the same as the number of its corresponding position in the reference sequence. For example, in a case where a variant has a deletion relative to an aligned reference sequence, there will be no amino acid in the variant that corresponds to a position in the reference sequence at the site of deletion.
- numbered with reference to refers to the numbering of the residues of a specified reference sequence when the given amino acid or polynucleotide sequence is compared to the reference sequence.
- An amino acid residue in a protein "corresponds" to a given residue when it occupies the same essential structural position within the protein as the given residue.
- residues corresponding to a specific position in a protein e.g., EGFR
- a protein e.g., EGFR
- identity and location of residues corresponding to specific positions of the protein are identified in other protein sequences aligning to the protein.
- a selected residue in a selected protein corresponds to glutamic acid at position 138 when the selected residue occupies the same essential spatial or other structural relationship as a glutamic acid at position 138.
- the position in the aligned selected protein aligning with glutamic acid 138 is the to correspond to glutamic acid 138.
- a three dimensional structural alignment can also be used, e.g., where the structure of the selected protein is aligned for maximum correspondence with the glutamic acid at position 138, and the overall structures compared.
- an amino acid that occupies the same essential position as glutamic acid 138 in the structural model is the residue to correspond to the glutamic acid 138 residue.
- Constantly modified variants applies to both amino acid and nucleic acid sequences. With respect to particular nucleic acid sequences, “conservatively modified variants” refers to those nucleic acids that encode identical or essentially identical amino acid sequences. Because of the degeneracy of the genetic code, a number of nucleic acid sequences will encode any given protein. For instance, the codons GCA, GCC, GCG and GCU all encode the amino acid alanine. Thus, at every position where an alanine is specified by a codon, the codon can be altered to any of the corresponding codons described without altering the encoded polypeptide. Such nucleic acid variations are "silent variations,” which are one species of conservatively modified variations.
- Every nucleic acid sequence herein which encodes a polypeptide also describes every possible silent variation of the nucleic acid.
- each codon in a nucleic acid except AUG, which is ordinarily the only codon for methionine, and TGG, which is ordinarily the only codon for tryptophan
- TGG which is ordinarily the only codon for tryptophan
- amino acid sequences one of skill will recognize that individual substitutions, deletions or additions to a nucleic acid, peptide, polypeptide, or protein sequence which alters, adds or deletes a single amino acid or a small percentage of amino acids in the encoded sequence is a "conservatively modified variant" where the alteration results in the substitution of an amino acid with a chemically similar amino acid. Conservative substitution tables providing functionally similar amino acids are well known in the art. Such conservatively modified variants are in addition to and do not exclude polymorphic variants, interspecies homologs, and alleles of the disclosure.
- nucleic acids or polypeptide sequences refer to two or more sequences or subsequences that are the same or have a specified percentage of amino acid residues or nucleotides that are the same (i.e., about 60% identity, preferably 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or higher identity over a specified region, when compared and aligned for maximum correspondence over a comparison window or designated region) as measured using a BLAST or BLAST 2.0 sequence comparison algorithms with default parameters described below, or by manual alignment and visual inspection (see, e.g., NCBI web site http://www.ncbi.nlm.nih.gov/BLAST/ or the like).
- sequences are then said to be “substantially identical.”
- This definition also refers to, or may be applied to, the compliment of a test sequence.
- the definition also includes sequences that have deletions and/or additions, as well as those that have substitutions.
- the preferred algorithms can account for gaps and the like.
- identity exists over a region that is at least about 25 amino acids or nucleotides in length, or more preferably over a region that is 50-100 amino acids or nucleotides in length.
- Percentage of sequence identity is determined by comparing two optimally aligned sequences over a comparison window, wherein the portion of the polynucleotide or polypeptide sequence in the comparison window may comprise additions or deletions (i.e., gaps) as compared to the reference sequence (which does not comprise additions or deletions) for optimal alignment of the two sequences. The percentage is calculated by determining the number of positions at which the identical nucleic acid base or amino acid residue occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the window of comparison and multiplying the result by 100 to yield the percentage of sequence identity.
- a “comparison window”, as used herein, includes reference to a segment of any one of the number of contiguous positions selected from the group consisting of, e.g., a full length sequence or from 20 to 600, about 50 to about 200, or about 100 to about 150 amino acids or nucleotides in which a sequence may be compared to a reference sequence of the same number of contiguous positions after the two sequences are optimally aligned.
- Methods of alignment of sequences for comparison are well-known in the art. Optimal alignment of sequences for comparison can be conducted, e.g., by the local homology algorithm of Smith and Waterman (1970) Adv. Appl. Math.
- HSPs high scoring sequence pairs
- Cumulative scores are calculated using, for nucleotide sequences, the parameters M (reward score for a pair of matching residues; always > 0) and N (penalty score for mismatching residues; always ⁇ 0).
- M forward score for a pair of matching residues; always > 0
- N penalty score for mismatching residues; always ⁇ 0.
- a scoring matrix is used to calculate the cumulative score. Extension of the word hits in each direction are halted when: the cumulative alignment score falls off by the quantity X from its maximum achieved value; the cumulative score goes to zero or below, due to the accumulation of one or more negative scoring residue alignments; or the end of either sequence is reached.
- the BLAST algorithm parameters W, T, and X determine the sensitivity and speed of the alignment.
- W word length
- E expectation
- B B- 50
- E expectation
- B B- 50
- E expectation
- the BLAST algorithm also performs a statistical analysis of the similarity between two sequences (see, e.g.. Karlin and Altschul (1993) Proc. Natl. Acad. Sci. USA 90:5873- 5787).
- One measure of similarity provided by the BLAST algorithm is the smallest sum probability (P(N)), which provides an indication of the probability by which a match between two nucleotide or amino acid sequences would occur by chance.
- P(N) the smallest sum probability
- a nucleic acid is considered similar to a reference sequence if the smallest sum probability in a comparison of the test nucleic acid to the reference nucleic acid is less than about 0.2, more preferably less than about 0.01, and most preferably less than about 0.001.
- nucleic acid sequences or polypeptides are substantially identical is that the polypeptide encoded by the first nucleic acid is immunologically cross reactive with the antibodies raised against the polypeptide encoded by the second nucleic acid, as described below.
- a polypeptide is typically substantially identical to a second polypeptide, for example, where the two peptides differ only by conservative substitutions.
- Another indication that two nucleic acid sequences are substantially identical is that the two molecules or their complements hybridize to each other under stringent conditions, as described below.
- Yet another indication that two nucleic acid sequences are substantially identical is that the same primers can be used to amplify the sequence.
- Antibodies are large, complex molecules (molecular weight of -150,000 or about 1320 amino acids) with intricate internal structure.
- a natural antibody molecule contains two identical pairs of polypeptide chains, each pair having one light chain and one heavy chain. Each light chain and heavy chain in turn consists of two regions: a variable (“V”) region, involved in binding the target antigen, and a constant (“C”) region that interacts with other components of the immune system.
- the light and heavy chain variable regions also referred to herein as light chain variable (VL) domain and heavy chain variable (VH) domain, respectively
- VL variable
- VH heavy chain variable domain
- CDRs complementarity determining regions
- the six CDRs in an antibody variable domain fold up together in 3- dimensional space to form the actual antibody binding site which docks onto the target antigen.
- the position and length of the CDRs have been precisely defined by Rabat, E. et al., Sequences of Proteins of Immunological Interest, U.S. Department of Health and Human Services, 1983, 1987.
- the part of a variable region not contained in the CDRs is called the framework (“FR”), which forms the environment for the CDRs.
- FR framework
- an “antibody variant” as provided herein refers to a polypeptide capable of binding to an antigen and including one or more structural domains (e.g., light chain variable domain, heavy chain variable domain) of an antibody or fragment thereof.
- Non-limiting examples of antibody variants include single-domain antibodies or nanobodies, monospecific Fab2, bispecific Fal3 ⁇ 4, trispecific Fab 3 , monovalent IgGs, scFv, bispecific antibodies, bispecific diabodies, trispecific triabodies, scFv-Fc, minibodies, IgNAR, V-NAR, hcIgG, VhH, or peptibodies.
- a “peptibody” as provided herein refers to a peptide moiety attached (through a covalent or non-covalent linker) to the Fc domain of an antibody.
- antibody variants known in the art include antibodies produced by cartilaginous fish or camelids. A general description of antibodies from camelids and the variable regions thereof and methods for their production, isolation, and use may be found in references WO97/49805 and WO 97/49805 which are incorporated by reference herein in their entirety and for all purposes. Likewise, antibodies from cartilaginous fish and the variable regions thereof and methods for their production, isolation, and use may be found in W02005/118629, which is incorporated by reference herein in its entirety and for all purposes.
- CDR LI complementarity determining regions
- CDR L2 complementarity determining regions
- CDR L3 complementarity determining regions
- the variable light chain provided herein includes in N-terminal to C-terminal direction a CDR LI, a CDR L2 and a CDR L3.
- CDR HI complementarity determining regions
- CDR H2 complementarity determining regions
- H3 variable heavy chain
- variable heavy chain provided herein includes in N-terminal to C-terminal direction a CDR HI, a CDR H2 and a CDR H3.
- the CDRs of the light chain are referred to as CDR1, CDR2, and CDR3 of VL and the CDRs of the heavy chain are referred to as CDR1, CDR2, and CDR3 of VH. See, for example the tables as provided herein.
- variable light chain includes in N-terminal to C-terminal direction a FR LI, a FR L2, a FR L3 and a FR L4.
- FR HI FR H2
- FR H3 FR H4
- FR H4 the variable heavy chain provided herein includes in N-terminal to C-terminal direction a FR HI, a FR H2, a FR H3 and a FR H4.
- An exemplary immunoglobulin (antibody) structural unit comprises a tetramer.
- Each tetramer is composed of two identical pairs of polypeptide chains, each pair having one “light” (about 25 kD) and one “heavy” chain (about 50-70 kD).
- the N-terminus of each chain defines a variable region of about 100 to 110 or more amino acids primarily responsible for antigen recognition.
- the terms variable light chain (VL), variable light chain (VL) domain or light chain variable region and variable heavy chain (VH), variable heavy chain (VH) domain or heavy chain variable region refer to these light and heavy chain regions, respectively.
- variable light chain (VL), variable light chain (VL) domain and light chain variable region as referred to herein may be used interchangeably.
- variable heavy chain (VH), variable heavy chain (VH) domain and heavy chain variable region as referred to herein may be used interchangeably.
- the Fc i.e. fragment crystallizable region
- the Fc region is the "base” or “tail” of an immunoglobulin and is typically composed of two heavy chains that contribute two or three constant domains depending on the class of the antibody. By binding to specific proteins, the Fc region ensures that each antibody generates an appropriate immune response for a given antigen.
- the Fc region also binds to various cell receptors, such as Fc receptors, and other immune molecules, such as complement proteins.
- light chain is used according to its ordinary meaning in the biological arts, and refers to the polypeptide formed by a light chain variable domain (VL) and a light chain constant domain (CL).
- VL light chain variable domain
- CL light chain constant domain
- heavy chain is used according to its ordinary meaning in the biological arts, and refers to the polypeptide formed by a heavy chain variable domain (VH) and one or more heavy chain constant domains (CHI, CH2, CH3).
- antibody is used according to its commonly known meaning in the art. Antibodies exist, e.g., as intact immunoglobulins or as a number of well-characterized fragments produced by digestion with various peptidases. Thus, for example, pepsin digests an antibody below the disulfide linkages in the hinge region to produce F(ab)' 2 , a dimer of Fab which itself is a light chain joined to VH-CHI by a disulfide bond. The F(ab)' 2 may be reduced under mild conditions to break the disulfide linkage in the hinge region, thereby converting the F(ab)' 2 dimer into an Fab' monomer.
- the Fab' monomer is essentially Fab with part of the hinge region (see Fundamental Immunology (Paul ed., 3d ed. 1993). While various antibody fragments are defined in terms of the digestion of an intact antibody, one of skill will appreciate that such fragments may be synthesized de novo either chemically or by using recombinant DNA methodology. Thus, the term antibody, as used herein, also includes antibody fragments either produced by the modification of whole antibodies, or those synthesized de novo using recombinant DNA methodologies (e.g., single chain Fv) or those identified using phage display libraries (see, e.g., McCafferty et al, Nature 348:552-554 (1990)).
- an antibody as referred to herein further includes antibody variants such as single domain antibodies.
- an antibody includes a single monomeric variable antibody domain.
- the antibody includes a variable light chain (VL) domain or a variable heavy chain (VH) domain.
- the antibody is a variable light chain (VL) domain or a variable heavy chain (VH) domain.
- mAb monoclonal or polyclonal antibodies
- Techniques for the production of single chain antibodies can be adapted to produce antibodies to polypeptides of this invention.
- transgenic mice, or other organisms such as other mammals may be used to express humanized antibodies.
- phage display technology can be used to identify antibodies and heteromeric Fab fragments that specifically bind to selected antigens (see, e.g.. McCafferty etal., Nature 348:552-554 (1990); Marks etal, Biotechnology 10:779-783 (1992)).
- a single-chain variable fragment is typically a fusion protein of the variable domains of the heavy (VH) and light chain (VL) of immunoglobulins, connected with a short linker peptide of 10 to about 25 amino acids.
- the linker may usually be rich in glycine for flexibility, as well as serine or threonine for solubility.
- the linker can either connect the N- terminus of the VH with the C-terminus of the VL, or vice versa.
- the epitope of a mAb is the region of its antigen to which the mAb binds.
- Two antibodies bind to the same or overlapping epitope if each competitively inhibits (blocks) binding of the other to the antigen. That is, a lx, 5x, lOx, 20x or lOOx excess of one antibody inhibits binding of the other by at least 30% but preferably 50%, 75%, 90% or even 99% as measured in a competitive binding assay (see, e.g., Junghans etal, Cancer Res. 50:1495, 1990).
- two antibodies have the same epitope if essentially all amino acid mutations in the antigen that reduce or eliminate binding of one antibody reduce or eliminate binding of the other.
- Two antibodies have overlapping epitopes if some amino acid mutations that reduce or eliminate binding of one antibody reduce or eliminate binding of the other.
- the genes encoding the heavy and light chains of an antibody of interest can be cloned from a cell, e.g., the genes encoding a monoclonal antibody can be cloned from a hybridoma and used to produce a recombinant monoclonal antibody.
- Gene libraries encoding heavy and light chains of monoclonal antibodies can also be made from hybridoma or plasma cells. Random combinations of the heavy and light chain gene products generate a large pool of antibodies with different antigenic specificity (see, e.g., Kuby, Immunology (3rd ed. 1997)).
- Techniques for the production of single chain antibodies or recombinant antibodies U.S. Patent 4,946,778, U.S. Patent No.
- transgenic mice or other organisms such as other mammals, may be used to express humanized or human antibodies (see, e.g., U.S. Patent Nos. 5,545,807; 5,545,806; 5,569,825; 5,625,126; 5,633,425; 5,661,016, Marks et al., Bio/Technology 10:779-783 (1992); Lonberg et al., Nature 368:856-859 (1994); Morrison, Nature 368:812-13 (1994); Fishwild et al., Nature Biotechnology 14:845-51 (1996); Neuberger, Nature Biotechnology 14:826 (1996); and Lonberg & Huszar, Intern.
- phage display technology can be used to identify antibodies and heteromeric Fab fragments that specifically bind to selected antigens (see, e.g., McCafferty et al., Nature 348:552-554 (1990); Marks et al., Biotechnology 10:779-783 (1992)).
- Antibodies can also be made bispecific, i.e., able to recognize two different antigens (see, e.g., WO 93/08829, Traunecker et al., EMBO J. 10:3655-3659 (1991); and Suresh et al., Methods in Enzymology 121:210 (1986)).
- Antibodies can also be heteroconjugates, e.g., two covalently joined antibodies, or immunotoxins (see, e.g., U.S. Patent No. 4,676,980 , WO 91/00360; WO 92/200373; and EP 03089).
- a humanized antibody has one or more amino acid residues introduced into it from a source which is non-human. These non-human amino acid residues are often referred to as import residues, which are typically taken from an import variable domain. Humanization can be essentially performed following the method of Winter and co workers (see, e.g., Morrison et al., PNAS USA, 81:6851-6855 (1984), Jones et al., Nature 321:522-525 (1986); Riechmann et al., Nature 332:323-327 (1988); Morrison and Oi, Adv. Immunol., 44:65-92 (1988), Verhoeyen et al., Science 239:1534-1536 (1988) and Presta,
- humanized antibodies are chimeric antibodies (U.S. Patent No. 4,816,567), wherein substantially less than an intact human variable domain has been substituted by the corresponding sequence from a non-human species.
- humanized antibodies are typically human antibodies in which some CDR residues and possibly some FR residues are substituted by residues from analogous sites in rodent antibodies.
- polynucleotides comprising a first sequence coding for humanized immunoglobulin framework regions and a second sequence set coding for the desired immunoglobulin complementarity determining regions can be produced synthetically or by combining appropriate cDNA and genomic DNA segments.
- Human constant region DNA sequences can be isolated in accordance with well known procedures from a variety of human cells.
- a "chimeric antibody” is an antibody molecule in which (a) the constant region, or a portion thereof, is altered, replaced or exchanged so that the antigen binding site (e.g, variable region including domain VH and VL) is linked to a constant region of a different or altered class, effector function and/or species, or an entirely different molecule which confers new properties to the chimeric antibody, e.g., an enzyme, toxin, hormone, growth factor, drug, etc.; or (b) the variable region, or a portion thereof, is altered, replaced or exchanged with a variable region having a different or altered antigen specificity.
- the preferred antibodies of, and for use according to the invention include humanized and/or chimeric monoclonal antibodies.
- the specified antibodies bind to a particular protein at least two times the background and more typically more than 10 to 100 times background.
- Specific binding to an antibody under such conditions requires an antibody that is selected for its specificity for a particular protein.
- polyclonal antibodies can be selected to obtain only a subset of antibodies that are specifically immunoreactive with the selected antigen and not with other proteins.
- a variety of immunoassay formats may be used to select antibodies specifically immunoreactive with a particular protein.
- solid-phase ELISA immunoassays are routinely used to select antibodies specifically immunoreactive with a protein (see, e.g., Harlow & Lane, Using Antibodies, A Laboratory Manual (1998) for a description of immunoassay formats and conditions that can be used to determine specific immunoreactivity).
- a "ligand” refers to an agent, e.g., a polypeptide or other molecule, capable of binding to a receptor or antibody, antibody variant, antibody region or fragment thereof.
- antibody- drug conjugate refers to a therapeutic agent conjugated or otherwise covalently bound to to an antibody.
- the named protein includes any of the protein’s naturally occurring forms, variants or homologs that maintain the protein transcription factor activity (e.g., within at least 50%, 80%, 90%, 95%, 96%, 97%, 98%, 99% or 100% activity compared to the native protein).
- variants or homologs have at least 90%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity across the whole sequence or a portion of the sequence (e.g. a 50, 100, 150 or 200 continuous amino acid portion) compared to a naturally occurring form.
- the protein is the protein as identified by its NCBI sequence reference.
- the protein is the protein as identified by its NCBI sequence reference, homolog or functional fragment thereof.
- EGFR protein or "EGFR” as used herein includes any of the recombinant or naturally-occurring forms of epidermal growth factor receptor, also known as Proto-oncogene c-ErbB-1, Receptor tyrosine-protein kinase erbB-1, ERBB, ERBBl, HER1, or variants or homologs thereof that maintain EGFR activity (e.g. within at least 50%, 80%, 90%, 95%, 96%, 97%, 98%, 99% or 100% activity compared to EGFR).
- epidermal growth factor receptor also known as Proto-oncogene c-ErbB-1, Receptor tyrosine-protein kinase erbB-1, ERBB, ERBBl, HER1, or variants or homologs thereof that maintain EGFR activity (e.g. within at least 50%, 80%, 90%, 95%, 96%, 97%, 98%, 99% or 100% activity compared to EGFR).
- the variants or homologs have at least 90%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity across the whole sequence or a portion of the sequence (e.g. a 50, 100, 150 or 200 continuous amino acid portion) compared to a naturally occurring EGFR protein.
- the EGFR protein is substantially identical to the protein identified by the UniProt reference number P00533 or a variant or homolog having substantial identity thereto.
- the term "gene” means the segment of DNA involved in producing a protein; it includes regions preceding and following the coding region (leader and trailer) as well as intervening sequences (introns) between individual coding segments (exons).
- the leader, the trailer as well as the introns include regulatory elements that are necessary during the transcription and the translation of a gene.
- a “protein gene product” is a protein expressed from a particular gene.
- plasmid refers to a nucleic acid molecule that encodes for genes and/or regulatory elements necessary for the expression of genes. Expression of a gene from a plasmid can occur in cis or in trans. If a gene is expressed in cis, the gene and the regulatory elements are encoded by the same plasmid. Expression in trans refers to the instance where the gene and the regulatory elements are encoded by separate plasmids.
- a vector may be any agent capable of delivering or maintaining nucleic acid in a host cell, and includes viral vectors (e.g.
- retroviral vectors lentiviral vectors, adenoviral vectors, or adeno-associated viral vectors
- plasmids naked nucleic acids, nucleic acids complexed with polypeptide or other molecules and nucleic acids immobilized onto solid phase particles.
- the appropriate DNA sequence may be inserted into the vector by a variety of procedures. In general, the DNA sequence is inserted into an appropriate restriction endonuclease site(s) by procedures known in the art. Such procedures and others are deemed to be within the scope of those skilled in the art. Transcription of the DNA encoding the polypeptides of the present invention by higher eukaryotes is increased by inserting an enhancer sequence into the vector.
- Enhancers are cis-acting elements of DNA, usually about from 10 to 300 by that act on a promoter to increase its transcription. Examples including the SV40 enhancer on the late side of the replication origin by 100 to 270, a cytomegalovirus early promoter enhancer, the polyoma enhancer on the late side of the replication origin, and adenovirus enhancers.
- transfection can be used interchangeably and are defined as a process of introducing a nucleic acid molecule or a protein to a cell.
- Nucleic acids are introduced to a cell using non-viral or viral-based methods.
- the nucleic acid molecules may be gene sequences encoding complete proteins or functional portions thereof.
- Non-viral methods of transfection include any appropriate transfection method that does not use viral DNA or viral particles as a delivery system to introduce the nucleic acid molecule into the cell.
- Exemplary non-viral transfection methods include calcium phosphate transfection, liposomal transfection, nucleofection, sonoporation, transfection through heat shock, magnetifection and electroporation.
- the nucleic acid molecules are introduced into a cell using electroporation following standard procedures well known in the art.
- any useful viral vector may be used in the methods described herein.
- viral vectors include, but are not limited to retroviral, adenoviral, lentiviral and adeno-associated viral vectors.
- the nucleic acid molecules are introduced into a cell using a retroviral vector following standard procedures well known in the art.
- the terms "transfection” or "transduction” also refer to introducing proteins into a cell from the external environment. Typically, transduction or transfection of a protein relies on attachment of a peptide or protein capable of crossing the cell membrane to the protein of interest. See, e.g., Ford et al. (2001) Gene Therapy 8: 1-4 and Prochiantz (2007) Nat. Methods 4: 119-20.
- a "label” or a “detectable moiety” is a composition detectable by spectroscopic, photochemical, biochemical, immunochemical, chemical, or other physical means.
- useful labels include 32P, fluorescent dyes, electron-dense reagents, enzymes (e.g., as commonly used in an ELISA), biotin, digoxigenin, or haptens and proteins or other entities which can be made detectable, e.g., by incorporating a radiolabel into a peptide or antibody specifically reactive with a target peptide.
- detectable moiety includes a composition, substance, element, or compound; or moiety thereof; detectable by appropriate means such as spectroscopic, photochemical, biochemical, immunochemical, chemical, magnetic resonance imaging, or other physical means.
- useful detectable agents include 18 F, 32 P, 33 P, 45 Ti, 47 Sc, 52 Fe, 59 Fe, 62 Cu, 64 Cu, 67 Cu, 67 Ga, 68 Ga, 77 As, 86 Y, 90 Y.
- fluorescent dyes include fluorescent dyes), electron-dense reagents, enzymes (e.g., as commonly used in an ELISA), biotin, digoxigenin, paramagnetic molecules, paramagnetic nanoparticles, ultrasmall superparamagnetic iron oxide (“USPIO”) nanoparticles, USPIO nanoparticle aggregates, superparamagnetic iron oxide (“SPIO”) nanoparticles, SPIO nanoparticle aggregates, monochrystalline iron oxide nanoparticles, monochrystalline iron oxide, nanoparticle contrast agents, liposomes or other delivery vehicles containing Gadolinium chelate (“Gd-chelate”) molecules, Gadolinium, radioisotopes, radionuclides (e.g.
- microbubbles e.g. including microbubble shells including albumin, galactose, lipid, and/or polymers; microbubble gas core including air, heavy gas(es), perfluorcarbon, nitrogen, octafluoropropane, perflexane lipid microsphere, perflutren, etc.
- iodinated contrast agents e.g.
- a detectable moiety is a monovalent detectable agent or a detectable agent capable of forming a bond with another composition.
- the agent may be reacted with another long-tailed reagent having a long tail with one or more chelating groups atached to the long tail for binding to these ions.
- the long tail may be a polymer such as a polylysine, polysaccharide, or other derivatized or derivatizable chain having pendant groups to which the metals or ions may be added for binding.
- chelating groups that may be used according to the disclosure include, but are not limited to, ethylenediaminetetraacetic acid (EDTA), diethylenetriaminepentaacetic acid (DTP A),
- the chelate is normally linked to the PSMA antibody or functional antibody fragment by a group, which enables the formation of a bond to the molecule with minimal loss of immunoreactivity and minimal aggregation and/or internal cross-linking.
- the same chelates, when complexed with non-radioactive metals, such as manganese, iron and gadolinium are useful for MRI, when used along with the antibodies and carriers described herein.
- Macrocyclic chelates such as NOTA, DOTA, and TETA are of use with a variety of metals and radiometals including, but not limited to, radionuclides of gallium, ytrium and copper, respectively.
- Other ring-type chelates such as macrocyclic polyethers, which are of interest for stably binding nuclides, such as 223 Ra for RAIT may be used.
- chelating moieties may be used to attach a PET imaging agent, such as an A1- 18 F complex, to a targeting molecule for use in PET analysis.
- Contacting is used in accordance with its plain ordinary meaning and refers to the process of allowing at least two distinct species (e.g. antibodies and antigens) to become sufficiently proximal to react, interact, or physically touch. It should be appreciated; however, that the resulting reaction product can be produced directly from a reaction between the added reagents or from an intermediate from one or more of the added reagents which can be produced in the reaction mixture.
- species e.g. antibodies and antigens
- contacting may include allowing two species to react, interact, or physically touch, wherein the two species may be, for example, a pharmaceutical composition as provided herein and a cell.
- contacting includes, for example, allowing a pharmaceutical composition as described herein to interact with a cell.
- a "cell” as used herein, refers to a cell carrying out metabolic or other function sufficient to preserve or replicate its genomic DNA.
- a cell can be identified by well-known methods in the art including, for example, presence of an intact membrane, staining by a particular dye, ability to produce progeny or, in the case of a gamete, ability to combine with a second gamete to produce a viable offspring.
- Cells may include prokaryotic and eukaryotic cells.
- Prokaryotic cells include but are not limited to bacteria.
- Eukaryotic cells include, but are not limited to, yeast cells and cells derived from plants and animals, for example mammalian, insect (e.g., spodoptera) and human cells.
- recombinant when used with reference, e.g., to a cell, nucleic acid, protein, or vector, indicates that the cell, nucleic acid, protein or vector, has been modified by the introduction of a heterologous nucleic acid or protein or the alteration of a native nucleic acid or protein, or that the cell is derived from a cell so modified.
- recombinant cells express genes that are not found within the native (non-recombinant) form of the cell or express native genes that are otherwise abnormally expressed, under expressed or not expressed at all.
- Transgenic cells and plants are those that express a heterologous gene or coding sequence, typically as a result of recombinant methods.
- nucleic acid or protein when applied to a nucleic acid or protein, denotes that the nucleic acid or protein is essentially free of other cellular components with which it is associated in the natural state. It can be, for example, in a homogeneous state and may be in either a dry or aqueous solution. Purity and homogeneity are typically determined using analytical chemistry techniques such as polyacrylamide gel electrophoresis or high performance liquid chromatography. A protein that is the predominant species present in a preparation is substantially purified.
- heterologous when used with reference to portions of a nucleic acid indicates that the nucleic acid comprises two or more subsequences that are not found in the same relationship to each other in nature.
- the nucleic acid is typically recombinantly produced, having two or more sequences from unrelated genes arranged to make a new functional nucleic acid, e.g., a promoter from one source and a coding region from another source.
- a heterologous protein indicates that the protein comprises two or more subsequences that are not found in the same relationship to each other in nature (e.g., a fusion protein).
- exogenous refers to a molecule or substance (e.g. , a compound, nucleic acid or protein) that originates from outside a given cell or organism.
- an "exogenous promoter” as referred to herein is a promoter that does not originate from the cell or organism it is expressed by.
- endogenous or endogenous promoter refers to a molecule or substance that is native to, or originates within, a given cell or organism.
- inhibition means negatively affecting (e.g., decreasing proliferation) or killing the cell.
- inhibition refers to reduction of a disease or symptoms of disease (e.g., cancer, cancer cell proliferation).
- inhibition includes, at least in part, partially or totally blocking stimulation, decreasing, preventing, or delaying activation, or inactivating, desensitizing, or down-regulating signal transduction or enzymatic activity or the amount of a protein (e.g. EGFR protein).
- an “inhibitor” is a compound or protein that inhibits a receptor or another protein, e.g.,, by binding, partially or totally blocking, decreasing, preventing, delaying, inactivating, desensitizing, or down-regulating activity (e.g., a receptor activity or a protein activity).
- the term “inhibition”, “inhibit”, “inhibiting” and the like in reference to a protein-inhibitor interaction means negatively affecting (e.g. decreasing) the activity or function of the protein (e.g. EGFR protein) relative to the activity or function of the protein in the absence of the inhibitor.
- inhibition means negatively affecting (e.g. decreasing) the concentration or levels of EGFR relative to the concentration or level of the protein in the absence of the inhibitor.
- inhibition refers to reduction of a disease or symptoms of disease.
- inhibition refers to a reduction in the activity of EGFR.
- inhibition includes, at least in part, partially or totally blocking stimulation, decreasing, preventing, or delaying activation, or inactivating, desensitizing, or down-regulating signal transduction or enzymatic activity or the amount of EGFR.
- inhibition refers to a reduction of activity of EGFR resulting from a direct interaction (e.g. an inhibitor binds to EGFR).
- inhibition refers to a reduction of activity of EGFR from an indirect interaction (e.g. an inhibitor binds to a protein that activates EGFR, thereby preventing target protein activation).
- the terms “inhibitor,” “repressor” or “antagonist” or “downregulator” interchangeably refer to a substance capable of detectably decreasing the expression or activity of a given gene or protein (e.g. EGFR protein).
- the antagonist can decrease EGFR expression or activity 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more in comparison to a control in the absence of the antagonist.
- EGFR expression or activity is 1.5-fold, 2-fold, 3-fold, 4-fold, 5-fold, 10-fold or lower than the expression or activity in the absence of the antagonist.
- expression includes any step involved in the production of the polypeptide including, but not limited to, transcription, post-transcriptional modification, translation, post-translational modification, and secretion. Expression can be detected using conventional techniques for detecting protein (e.g., ELISA, Western blotting, flow cytometry, immunofluorescence, immunohistochemistry, etc.).
- Bio sample refers to materials obtained from or derived from a subject or patient.
- a biological sample includes sections of tissues such as biopsy and autopsy samples, and frozen sections taken for histological purposes.
- samples include bodily fluids such as blood and blood fractions or products (e.g., serum, plasma, platelets, red blood cells, and the like), sputum, tissue, cultured cells (e.g., primary cultures, explants, and transformed cells) stool, urine, synovial fluid, joint tissue, immune cells, hematopoietic cells, fibroblasts, macrophages, T cells, etc.
- a biological sample is typically obtained from a eukaryotic organism, such as a mammal such as a primate e.g., chimpanzee or human; cow; dog; cat; a rodent, e.g., guinea pig, rat, mouse; rabbit; or a bird; reptile; or fish.
- a mammal such as a primate e.g., chimpanzee or human; cow; dog; cat; a rodent, e.g., guinea pig, rat, mouse; rabbit; or a bird; reptile; or fish.
- a “control” or “standard control” refers to a sample, measurement, or value that serves as a reference, usually a known reference, for comparison to a test sample, measurement, or value.
- a test sample can be taken from a patient suspected of having a given disease (e.g. cancer) and compared to a known normal (non-dis eased) individual (e.g. a standard control subject).
- a standard control can also represent an average measurement or value gathered from a population of similar individuals (e.g. standard control subjects) that do not have a given disease (i.e. standard control population), e.g., healthy individuals with a similar medical background, same age, weight, etc.
- a standard control value can also be obtained from the same individual, e.g. from an earlier-obtained sample from the patient prior to disease onset.
- a control can be devised to compare therapeutic benefit based on pharmacological data (e.g. , half-life) or therapeutic measures (e.g., comparison of side effects). Controls are also valuable for determining the significance of data. For example, if values for a given parameter are widely variant in controls, variation in test samples will not be considered as significant.
- standard controls can be designed for assessment of any number of parameters (e.g. RNA levels, protein levels, specific cell types, specific bodily fluids, specific tissues, etc).
- Standard controls are also valuable for determining the significance (e.g. statistical significance) of data. For example, if values for a given parameter are widely variant in standard controls, variation in test samples will not be considered as significant.
- “Patient” or “subject in need thereof’ refers to a living organism suffering from or prone to a disease or condition that can be treated by administration of a composition or pharmaceutical composition as provided herein.
- Non-limiting examples include humans, other mammals, bovines, rats, mice, dogs, monkeys, goat, sheep, cows, deer, and other non-mammalian animals.
- a patient is human.
- the terms “disease” or “condition” refer to a state of being or health status of a patient or subject capable of being treated with the compounds or methods provided herein.
- the disease may be a cancer.
- the cancer may refer to a solid tumor malignancy.
- Solid tumor malignancies include malignant tumors that may be devoid of fluids or cysts.
- the solid tumor malignancy may include breast cancer, ovarian cancer, pancreatic cancer, cervical cancer, gastric cancer, renal cancer, head and neck cancer, bone cancer, skm cancer or prostate cancer.
- cancer refers to human cancers and carcinomas, sarcomas, adenocarcinomas, lymphomas, leukemias, including solid and lymphoid cancers, kidney, breast, lung, bladder, colon, ovarian, prostate, pancreas, stomach, brain, head and neck, skin, uterine, testicular, glioma, esophagus, and liver cancer, including hepatocarcinoma, lymphoma, including B-acute lymphoblastic lymphoma, non-Hodgkin’s lymphomas (e.g., Burkitt’s, Small Cell, and Large Cell lymphomas), Hodgkin’s lymphoma, leukemia (including acute myeloid leukemia (AML), ALL, and CML), or multiple myeloma.
- AML acute myeloid leukemia
- ALL acute myeloid leukemia
- CML multiple myeloma
- cancer refers to all types of cancer, neoplasm or malignant tumors found in mammals (e.g., humans), including leukemia, carcinomas and sarcomas.
- exemplary cancers that may be treated with a compound or method provided herein include breast cancer, colon cancer, kidney cancer, leukemia, lung cancer, melanoma, ovarian cancer,
- modulate is used in accordance with its plain ordinary meaning and refers to the act of changing or varying one or more properties. “Modulation” refers to the process of changing or varying one or more properties. For example, as applied to the effects of a modulator on a target protein, to modulate means to change by increasing or decreasing a property or function of the target molecule or the amount of the target molecule.
- a disease e.g. a protein associated disease, a cancer (e.g., breast cancer, lung cancer)
- the disease e.g. cancer
- a symptom of the disease is caused by (in whole or in part) the substance or substance activity or function.
- a causative agent could be a target for treatment of the disease.
- aberrant refers to different from normal. When used to describe enzymatic activity, aberrant refers to activity that is greater or less than a normal control or the average of normal non-diseased control samples. Aberrant activity may refer to an amount of activity that results in a disease, wherein returning the aberrant activity to a normal or non-disease-associated amount (e.g. by using a method as described herein), results in reduction of the disease or one or more disease symptoms.
- a "therapeutic agent” as referred to herein, is a composition useful in treating or preventing a disease such as cancer (e.g., leukemia).
- the therpaeutic agent is an anti-cancer agent.
- Anti-cancer agent is used in accordance with its plain ordinary meaning and refers to a composition (e.g. compound, drug, antagonist, inhibitor, modulator) having antineoplastic properties or the ability to inhibit the growth or proliferation of cells.
- an anti-cancer agent is a chemotherapeutic.
- an anti-cancer agent is an agent identified herein having utility in methods of treating cancer.
- an anti-cancer agent is an agent approved by the FDA or similar regulatory agency of a country other than the USA, for treating cancer.
- treating or “treatment of’ a condition, disease or disorder or symptoms associated with a condition, disease or disorder refers to an approach for obtaining beneficial or desired results, including clinical results.
- beneficial or desired clinical results can include, but are not limited to, alleviation or amelioration of one or more symptoms or conditions, diminishment of extent of condition, disorder or disease, stabilization of the state of condition, disorder or disease, prevention of development of condition, disorder or disease, prevention of spread of condition, disorder or disease, delay or slowing of condition, disorder or disease progression, delay or slowing of condition, disorder or disease onset, amelioration or palliation of the condition, disorder or disease state, and remission, whether partial or total.
- Treating can also mean prolonging survival of a subject beyond that expected in the absence of treatment. “Treating” can also mean inhibiting the progression of the condition, disorder or disease, slowing the progression of the condition, disorder or disease temporarily, although in some instances, it involves halting the progression of the condition, disorder or disease permanently.
- treatment, treat, or treating refers to a method of reducing the effects of one or more symptoms of a disease or condition characterized by expression of the protease or symptom of the disease or condition characterized by expression of the protease.
- treatment can refer to a 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100% reduction in the severity of an established disease, condition, or symptom of the disease or condition.
- a method for treating a disease is considered to be a treatment if there is a 10% reduction in one or more symptoms of the disease in a subject as compared to a control.
- the reduction can be a 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or any percent reduction in between 10% and 100% as compared to native or control levels.
- treatment does not necessarily refer to a cure or complete ablation of the disease, condition, or symptoms of the disease or condition.
- references to decreasing, reducing, or inhibiting include a change of 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%,
- dose refers to the amount of active ingredient given to an individual at each administration.
- the dose will vary depending on a number of factors, including the range of normal doses for a given therapy, frequency of administration; size and tolerance of the individual; severity of the condition; risk of side effects; and the route of administration.
- dose form refers to the particular format of the pharmaceutical or pharmaceutical composition, and depends on the route of administration.
- a dosage form can be in a liquid form for nebulization, e.g., for inhalants, in a tablet or liquid, e.g., for oral delivery, or a saline solution, e.g., for injection.
- terapéuticaally effective dose or amount as used herein is meant a dose that produces effects for which it is administered (e.g. treating or preventing a disease).
- dose and formulation will depend on the purpose of the treatment, and will be ascertainable by one skilled in the art using known techniques (see, e.g., Lieberman, Pharmaceutical Dosage Forms (vols. 1-3, 1992); Lloyd, The Art, Science and Technology of Pharmaceutical Compounding (1999); Remington: The Science and Practice of Pharmacy, 20th Edition, Gennaro, Editor (2003), and Pickar, Dosage Calculations (1999)).
- a therapeutically effective amount will show an increase or decrease of at least 5%, 10%, 15%, 20%, 25%, 40%, 50%, 60%, 75%, 80%, 90%, or at least 100%.
- Therapeutic efficacy can also be expressed as “-fold” increase or decrease.
- a therapeutically effective amount can have at least a 1.2-fold, 1.5 -fold, 2-fold, 5 -fold, or more effect over a standard control.
- a therapeutically effective dose or amount may ameliorate one or more symptoms of a disease.
- a therapeutically effective dose or amount may prevent or delay the onset of a disease or one or more symptoms of a disease when the effect for which it is being administered is to treat a person who is at risk of developing the disease.
- administering means oral administration, administration as a suppository, topical contact, intravenous, intraperitoneal, intramuscular, intralesional, intrathecal, intranasal or subcutaneous administration, or the implantation of a slow-release device, e.g., a mini-osmotic pump, to a subject.
- Administration is by any route, including parenteral and transmucosal (e.g., buccal, sublingual, palatal, gingival, nasal, vaginal, rectal, or transdermal).
- Parenteral administration includes, e.g., intravenous, intramuscular, intra arteriole, intradermal, subcutaneous, intraperitoneal, intraventricular, and intracranial.
- Other modes of delivery include, but are not limited to, the use of liposomal formulations, intravenous infusion, transdermal patches, etc.
- co-administer it is meant that a composition described herein is administered at the same time, just prior to, or just after the administration of one or more additional therapies, for example cancer therapies such as chemotherapy, hormonal therapy, radiotherapy, or immunotherapy.
- the compounds of the invention can be administered alone or can be coadministered to the patient.
- Coadministration is meant to include simultaneous or sequential administration of the compounds individually or in combination (more than one compound).
- the preparations can also be combined, when desired, with other active substances (e.g. to reduce metabolic degradation).
- the compositions of the present invention can be delivered by transdermally, by a topical route, formulated as applicator sticks, solutions, suspensions, emulsions, gels, creams, ointments, pastes, jellies, paints, powders, and aerosols.
- compositions of the present invention may additionally include components to provide sustained release and/or comfort.
- Such components include high molecular weight, anionic mucomimetic polymers, gelling polysaccharides and finely-divided drug carrier substrates. These components are discussed in greater detail in U.S. Pat. Nos. 4,911,920; 5,403,841; 5,212,162; and 4,861,760. The entire contents of these patents are incorporated herein by reference in their entirety for all purposes.
- the compositions of the present invention can also be delivered as microspheres for slow release in the body.
- microspheres can be administered via intradermal injection of drug-containing microspheres, which slowly release subcutaneously (see Rao, J. Biomater Sci. Polym. Ed.
- the formulations of the compositions of the present invention can be delivered by the use of liposomes which fuse with the cellular membrane or are endocytosed, i.e., by employing receptor ligands attached to the liposome, that bind to surface membrane protein receptors of the cell resulting in endocytosis.
- liposomes particularly where the liposome surface carries receptor ligands specific for target cells, or are otherwise preferentially directed to a specific organ, one can focus the delivery of the compositions of the present invention into the target cells in vivo.
- liposomes particularly where the liposome surface carries receptor ligands specific for target cells, or are otherwise preferentially directed to a specific organ.
- compositions of the present invention can also be delivered as nanoparticles.
- composition will generally comprise agents for buffering and preservation in storage, and can include buffers and carriers for appropriate delivery, depending on the route of administration.
- “Pharmaceutically acceptable excipient” and “pharmaceutically acceptable carrier” refer to a substance that aids the administration of an active agent to and absorption by a subject and can be included in the compositions of the present invention without causing a significant adverse toxicological effect on the patient.
- Non-limiting examples of pharmaceutically acceptable excipients include water, NaCl, normal saline solutions, lactated Ringer’s, normal sucrose, normal glucose, binders, fillers, disintegrants, lubricants, coatings, sweeteners, flavors, salt solutions (such as Ringer's solution), alcohols, oils, gelatins, carbohydrates such as lactose, amylose or starch, fatty acid esters, hydroxymethy cellulose, polyvinyl pyrrolidine, and colors, and the like.
- Such preparations can be sterilized and, if desired, mixed with auxiliary agents such as lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, coloring, and/or aromatic substances and the like that do not deleteriously react with the compounds of the invention.
- auxiliary agents such as lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, coloring, and/or aromatic substances and the like that do not deleteriously react with the compounds of the invention.
- auxiliary agents such as lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, coloring, and/or aromatic substances and the like that do not deleteriously react with the compounds of the invention.
- auxiliary agents such as lubricants, preservatives, stabilizers, wetting agents
- pharmaceutically acceptable salt refers to salts derived from a variety of organic and inorganic counter ions well known in the art and include, by way of example only, sodium, potassium, calcium, magnesium, ammonium, tetraalkylammonium, and the like; and when the molecule contains a basic functionality, salts of organic or inorganic acids, such as hydrochloride, hydrobromide, tartrate, mesylate, acetate, maleate, oxalate and the like.
- preparation is intended to include the formulation of the active compound with encapsulating material as a carrier providing a capsule in which the active component with or without other carriers, is surrounded by a carrier, which is thus in association with it.
- a carrier which is thus in association with it.
- cachets and lozenges are included. Tablets, powders, capsules, pills, cachets, and lozenges can be used as solid dosage forms suitable for oral administration.
- the pharmaceutical preparation is optionally in unit dosage form.
- the preparation is subdivided into unit doses containing appropriate quantities of the active component.
- the unit dosage form can be a packaged preparation, the package containing discrete quantities of preparation, such as packeted tablets, capsules, and powders in vials or ampoules.
- the unit dosage form can be a capsule, tablet, cachet, or lozenge itself, or it can be the appropriate number of any of these in packaged form.
- the unit dosage form can be of a frozen dispersion.
- genetic modification means any process that adds, deletes, alters, or disrupts an endogenous nucleotide sequence and includes, but is not limited to viral mediated gene transfer, liposome mediated transfer, transformation, transfection and transduction, e.g., viral mediated gene transfer such as the use of vectors based on DNA viruses such as lentivirus, adenovirus, retroviruses, adeno-associated virus and herpes virus.
- Variant refers to polypeptides having amino acid sequences that differ to some extent from a native sequence polypeptide. Ordinarily, amino acid sequence variants will possess at least about 80% sequence identity, more preferably, at least about 90% homologous by sequence. The amino acid sequence variants may possess substitutions, deletions, and/or insertions at certain positions within the reference amino acid sequence.
- ADCC antibody-dependent cell-mediated cytotoxicity
- Fc receptors such as natural killer cells, neutrophils, and macrophages
- ADCC activity may be assessed using methods, such as those described in U.S. Pat. No. 5,821,337.
- effector cells are leukocytes which express one or more effector cell ligands (.e.g, constant region receptors such as CD16) and perform effector functions.
- effector cell ligand refers to a cell surface molecule expressed on an effector cell of the immune system (e.g., a cytotoxic T cell, a helper T cell, a B cell, a natural killer cell). Upon binding of the antibody to the effector cell ligand expressed on the effector cell, the effector cell is activated and able to exert its function (e.g., selective killing or eradication of malignant, infected or otherwise unhealthy cells).
- the effector cell ligand is a CD3 protein.
- the effector cell ligand is a CD16 protein.
- the effector cell ligand is a CD32 protein.
- the effector cell ligand is a NKp46 protein.
- Non-immunogenic refers to a material that does not initiate, provoke or enhance an immune response where the immune response includes the adaptive and/or innate immune responses.
- Receptor means a polypeptide that is capable of specific binding to a molecule. Whereas many receptors may typically operate on the surface of a cell, some receptors may bind ligands when located inside the cell (and prior to transport to the surface) or may reside predominantly intra-cellularly and bind ligand therein.
- truncated EGFR or “tEGFR” as used herein refers to an EGFR protein that includes EGFR domain IV, and does not include the membrane distal EGF -binding domains I, II or III or the cytoplasmic signaling tail. In embodiments, the tEGFR does not include EGFR domain I, EGFR domain II, EGFR domain II and the cytoplasmic signaling tail.
- EGFR domain IV refers to the amino acid sequence typically found between domain III and the transmembrane domain of recombinant or naturally occurring forms of EGFR protein.
- EGFR domain IV, or variants or homologs thereof have at least 90%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity across the whole sequence or a portion of the sequence (e.g. a 50, 100, 150 or 200 continuous amino acid portion) compared to a naturally occurring EGFR domain IV.
- the domain IV EGFR is substantially identical to the domain IV of the protein identified by the UniProt reference number P00533 or a variant or homolog having substantial identity thereto.
- the domain IV EGFR has at least 90%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity across the whole sequence or a portion of the sequence (e.g. a 50, 100, 150 or 200 continuous amino acid portion) of the sequence of SEQ ID NO:276.
- the EGFR domain IV is substantially identical to the amino acid sequence identified by SEQ ID NO:276.
- EGFR domain III or “domain III EGFR” refers to the amino acid sequence typically found between domain II EGFR and domain IV EGFR of recombinant or naturally occurring forms of EGFR protein.
- EGFR domain III, or variants or homologs thereof have at least 90%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity across the whole sequence or a portion of the sequence (e.g. a 50, 100, 150 or 200 continuous amino acid portion) compared to a naturally occurring EGFR domain III.
- chimeric antigen receptor is used according to its conventional meaning in the art refers to a recombinant protein including an antibody region and a transmembrane region.
- an "antibody region” as provided herein refers to a monovalent or multivalent protein moiety that forms part of an antibody. A person of ordinary skill in the art would therefor immediately recognize that the antibody region is a protein moiety capable of binding an antigen (epitope). Thus, the antibody region provided herein may include a domain of an antibody or fragment (e.g., Fab) thereof. In embodiments, the antibody region includes a variable light chain domain and a variable heavy chain domain.
- a “variable light chain domain” as provided herein refers to a polypeptide including a light chain variable (VL) region. In embodiments, the variable light chain domain is a light chain variable (VL) region.
- a “variable heavy chain domain” as provided herein refers to a polypeptide including a heavy chain variable (VH) region. In embodiments, the variable heavy chain domain is a heavy chain variable (VH) region.
- a “transmembrane domain” as provided herein refers to a polypeptide forming part of a biological membrane.
- the transmembrane domain provided herein is capable of spanning a biological membrane (e.g., a cellular membrane) from one side of the membrane through to the other side of the membrane.
- the transmembrane domain spans from the intracellular side to the extracellular side of a cellular membrane.
- Transmembrane domains may include non-polar, hydrophobic residues, which anchor the proteins provided herein including embodiments thereof in a biological membrane (e.g., cellular membrane of a T cell). Any transmembrane domain capable of anchoring the proteins provided herein including embodiments thereof are contemplated.
- Non-limiting examples of transmembrane domains include the transmembrane domains of CD28, CD8, CD4 or CD3-zeta.
- the chimeric antigen receptor further includes an intracellular T- cell signaling domain.
- An "intracellular T-cell signaling domain" as provided herein includes amino acid sequences capable of providing primary signaling in response to binding of an antigen to the antibody region provided herein including embodiments thereof.
- the signaling of the intracellular T-cell signaling domain results in activation of the T cell expressing the same.
- the signaling of the intracellular T-cell signaling domain results in proliferation (cell division) of the T cell expressing the same.
- the signaling of the intracellular T-cell signaling domain results expression by said T cell of proteins known in the art to characteristic of activated T cell (e.g., CTLA-4, PD-1, CD28, CD69).
- the intracellular T-cell signaling domain is a CD3 z intracellular T-cell signaling domain.
- cancer antigen refers to a molecule expressed on a cancer cell.
- the cancer antigen is expressed at a higher level relative to a standard control.
- the cancer antigen is expressed on a healthy cell.
- a “standard control” can be the level of expression of the cancer antigen of a healthy cell.
- the standard control may be the expression level of the cancer antigen in a cell from a healthy subject (i.e. a subject that does not have cancer).
- the standard control may be the expression level of a non- cancerous cell derived from the same subject as the cancer antigen expressing cancer.
- the standard control is an expression level of a low cancer antigen or cancer antigen negative cancer cell.
- the standard control can be the expression level of a biological sample comprising healthy cells (i.e. non-cancer cells).
- the standard control can be the expression level of cells from a subject that has already been treated for a cancer antigen expressing cancer. In instances, the control value can be obtained from the same subject (i.e. from a later-obtained sample, subsequent to treatment of the cancer antigen expressing cancer).
- the standard control can also represent an average expression level gathered from a population of similar subjects (i.e. healthy individuals with a similar medical background, same age, weight, etc.).
- the expression level of a cancer antigen is at least 2-fold higher than the expression level of a standard control.
- the expression level of a cancer antigen is at least 5, 10, 50, 100, 150, 200,
- the expression level of a cancer antigen is 5, 10, 50, 100, 200, 300, 400, 500, 1,000, 10,000 or 100,000-fold higher than the expression level of a standard control.
- CD 19 protein or “CD 19” as used herein includes any of the recombinant or naturally-occurring forms of B-lymphocyte antigen CD 19, also known as CD 19 molecule (Cluster of Differentiation 19), B-Lymphocyte Surface Antigen B4, T-Cell Surface Antigen Leu- 12 and CVID3, or variants or homologs thereof that maintain CD 19 activity (e.g. within at least 50%, 80%, 90%, 95%, 96%, 97%, 98%, 99% or 100% activity compared to CD 19).
- the variants or homologs have at least 90%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity across the whole sequence or a portion of the sequence (e.g. a 50, 100, 150 or 200 continuous amino acid portion) compared to a naturally occurring CD 19 protein.
- the CD 19 protein is substantially identical to the protein identified by the UniProt reference number P15391 or a variant or homolog having substantial identity thereto.
- IL-15 protein or "IL-15” as used herein includes any of the recombinant or naturally-occurring forms of Interleukin- 15 (IL-15), or variants or homologs thereof that maintain IL-15 activity (e.g. within at least 50%, 80%, 90%, 95%, 96%, 97%, 98%, 99% or 100% activity compared to IL-15).
- the variants or homologs have at least 90%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity across the whole sequence or a portion of the sequence (e.g. a 50, 100, 150 or 200 continuous amino acid portion) compared to a naturally occurring IL-15 protein.
- the IL-15 protein is substantially identical to the protein identified by the UniProt reference number P40933 or a variant or homolog having substantial identity thereto.
- self-cleaving peptidyl sequence refers to a class of peptide sequences that can induce ribosomal skipping during translation, which results in the generation of multiple peptides originally encoded by a single mRNA.
- the self-cleaving peptidyl sequence is a T2A sequence.
- the self-cleaving peptidyl sequence is a T2A sequence or a 2A sequence.
- the self-cleaving peptidyl sequence is a foot-and-mouth disease virus sequence.
- the self cleaving peptidyl sequence is PVKQLLNFDLLKLAGDVESNPGP.
- the self-cleaving peptidyl sequence is an equine rhinitis A virus sequence. In embodiments, the self-cleaving peptidyl sequence is QCTNYALLKLAGDVESNPGP. In embodiments, the self-cleaving peptidyl sequence is a porcine teschovirus 1 sequence. In embodiments, the self-cleaving peptidyl sequence is ATNFSLLKQAGDVEENPGP. In embodiments, the self cleaving peptidyl sequence is Thosea asigna virus sequence. In embodiments, the self cleaving peptidyl sequence is EGRGSLLTCGDVESNPGP.
- a gene encoding a modified endogenous cell-surface molecule that may be used as a non-immunogenic selection epitope compatible with immunomagnetic selection.
- a non-immunogenic selection epitope may facilitate immunotherapy in cancer patients without undesirable immunologic rejection of cell products.
- the endogenous cell surface molecule e.g., tEGFR
- the endogenous cell surface molecule may be modified or truncated to retain an extracellular epitope recognized by a known antibody or functional fragment thereof, and to remove any signaling or trafficking domains and/or any extracellular domains unrecognized by said known antibody.
- a modified endogenous cell surface molecule e.g., tEGFR which lacks a signaling or trafficking domain and/or any extracellular domains unrecognized by said known antibody is rendered inert.
- the modified endogenous cell-surface molecule may be, but is not limited to, any non-immunogenic cell-surface related receptor, glycoprotein, cell adhesion molecule, antigen, integrin or cluster of differentiation (CD) that is modified as described herein. Modification of such cell-surface molecules is accomplished by keeping an epitope that is recognized by a known antibody or functional fragment thereof; and removing any signaling or trafficking domains and/or any extracellular domains unrecognized by a known antibody. Removal of the signaling or trafficking domains and/or any extracellular domains unrecognized by a known antibody renders the endogenous cell-surface molecule non- immunogenic and/or inert.
- CD cluster of differentiation
- endogenous cell-surface molecules that may be modified or truncated according to the embodiments described herein include, but are not limited to EpCAM, VEGFR, integrals (e.g., integrals . alpha.. nu... beta.3, .alpha.4, . alpha.. PI.b.beta.3,
- TNF receptor superfamily e.g., TRAIL-R1, TRAIL-R2
- PDGF Receptor interferon receptor
- folate receptor GPNMB
- ICAM-1 ICAM-1
- HLA-DR CEA
- CA-125 MUC1
- TAG-72 IL-6 receptor
- 5T4 GD2, GD3, or clusters of differentiation e.g., CD2, CD3, CD4, CD5, CD11, CDlla/LFA-1, CD15, CD18/ITGB2, CD19, CD20, CD22, CD23/IgE Receptor, CD25, CD28, CD30, CD33, CD38, CD40, CD41, CD44, CD51, CD52, CD62L, CD74, CD80, CD125, CD147/basigin,
- Corresponding antibodies that may be used to recognize a modified or truncated endogenous cell-surface molecule include any of the antibodies provided herein including embodiments thereof.
- the modified endogenous cell-surface molecule e.g., tEGFR
- tEGFR tyrosine kinase receptor gene
- tyrosine kinase receptors that may be modified or truncated according to the embodiments described herein include, but are not limited to, members of the endothelial growth factor receptor family (EGRF/ErbB 1/HERl ; ErbB2/HER2/neu; ErbB3/HER3; ErbB4/HER4), hepatocyte growth factor receptor (HGFR/c-MET) and insulin-like growth factor receptor- 1 (IGF-1R).
- modified tyrosine kinase receptors retain an extracellular epitope recognized by a known antibody or functional fragment thereof, and lack at least a tyrosine kinase domain.
- a modified tyrosine kinase receptor which lacks at least a tyrosine kinase domain renders the receptor inert.
- the modified endogenous cell surface molecule is a truncated EGFR (tEGFR) that includes an EGFR domain IV and does not include an EGFR domain III.
- tEGFR does not include Domain I, Domain II, Domain III, the Juxtamembrane Domain and the Tyrosine Kinase Domain as compared to an unmodified EGFR.
- a modified endogenous cell-surface molecule may be used as a marker for in vivo T cell engraftment.
- the modified endogenous cell- surface molecule is EGFRt
- the EGFRt may be used to track the uptake of the T cells to which it is attached in vivo without affecting cellular function of the T cells or the cells to which the T cells are targeted, such as bone marrow cells in a transplant situation.
- the use of an anti-domain IV EGFR antibody as provided herein and conjugated to probes or reporter genes such as sr39TK may be used to improve the tracking potential of EGFRt-expressing cells to patients via PET imaging techniques.
- a modified endogenous cell-surface molecule may be used to induce cell suicide.
- EGFRt may be used as a suicide gene via anti domain IV EGFR antibody -mediated complement and/or antibody dependent cell mediated cytotoxicity (ADCC) pathways.
- ADCC antibody dependent cell mediated cytotoxicity
- the truncated epidermal growth factor receptor (EGFRt) selection epitope or other modified cell-surface molecule is attached to other sequences.
- One exemplary sequence is the GMCSFR alpha chain signal sequence, which directs surface expression, attached to EGFRt.
- GMCSFR is encoded by nucleotides 1-66 and EGFRt is encoded by nucleotides 67-1071. See FIG. 29. Also in FIG. 29 is the antisense strand and amino acid sequences of GMCSFR alpha chain signal sequence linked to EGFRt.
- Another such sequence is a codon-optimized cDNA sequence encoding an anti-CD 19 costimulatory chimeric antigen receptor (CD19R-CD28gg-Zeta(CO)), and a cleavable T2A linker.
- Cytotoxic T lymphocytes (CTLs) modified to express a CD19-specific chimeric antigen receptor (CAR) that signals via a cytoplasmic costimulatory (CD28) domain fused to the cytoplasmic CD3-.zeta. domain exhibits superior anti-tumor potency that can be attributed to CD28-mediated survival and enhanced cytokine production.
- This construct may be further modified to incorporate a C-terminal 2A cleavable linker followed by the coding sequence for a truncated human EGFR (EGFRt) for the purpose of immunomagnetic purification of CAR-expressing transductants using anti-domain IV EGFR antibody-biotin/anti-biotin microbeads.
- EGFRt truncated human EGFR
- FIG. 30 See the CD19R-CD28gg-Zeta(CO)-T2A-EGFRt sequence attached as FIG. 30, (nucleotide sense strand), (nucleotide anti-sense strand), and (protein).
- Lentivector transduction of primary human T cells with this codon-optimized cDNA directs the coordinated expression of the CAR and EGFRt (FIG. 30).
- EGFRt is a truncated human epidermal growth factor receptor that lacks the membrane distal EGF- binding domain and the ectoplasmic signaling tail, but retains the extracellular membrane proximal epitope recognized by the anti-domain IV EGFR antibody provided herein including embodiments thereof.
- Biotinylated-anti-domain IV EGFR antibody is applied to immunomagnetic selection in combination with anti-biotin microbeads (Miltenyi).
- Human OKT3 blasts that had been lentivirally transduced with CD19R-CD28gg-Zeta(CO)-T2A- EGFRt were subjected to immunomagnetic selection using the Miltenyi AutoMACS device, and the frequency of EGFRt+CAR+ T cells was enriched from 22% (pre-selection) to 99% (post-selection) without observable toxicity to the cell preparation.
- the EGFRt can be purified using fluorescence-based cell sorting techniques.
- EGFRt Due to the absence of the EGF-binding domains and intracellular signaling domains, EGFRt is inactive when expressed by T cells. Importantly, the EGFRt-selected T cells maintain their desired effector phenotype— including anti-tumor cyotoxic activity mediated by the chimeric antigen receptor that is coordinately expressed with the EGFRt— and remain amenable to established expansion protocols.
- this EGFRt has various advantages for immunotherapeutic cell products compared to other selection markers that have been previously reported. Specifically, unlike truncated CD4 and CD19, it is not endogenously expressed by subpopulations of lymphocytes. Furthermore, in contrast to truncated CD34 and low affinity nerve growth factor receptor, it does not have any activity that might negatively affect the immune cell product (i.e., in terms of signaling or trafficking). Lastly, it alone can be bound/recognized by any of the anti-domain IV EGFR antibodies provided herein including embodiments thereof. Together, these attributes make the EGFRt provided herein a superior selection marker for any transfection/transduction system that can be applied to the generation of cell products for adoptive immunotherapy. Thus, EGFRt is well suited to be used as a selection marker for, e.g., lentivirally transduced T cells of immunotherapeutic relevance.
- the immunoglobulin and the Fab of the antibodies provided herein bind domain IV of EGFR with differential affinity.
- the Fab of an antibody provided herein binds domain IV EGFR with lower affinity than the IgG of the same antibody. Therefore, and without being bound to any specific theory, the antibodies provided herein may be capable of selectively binding EGFR high expressing cells (e.g., cancer cells), thereby providing for highly specific antibody therapeutics without adverse effects.
- EGFR antibodies capable of binding truncated domain IV EGFR but substantially not binding to full-length EGFR.
- full-length EGFR refers to endogenous EGFR, which includes domain I, domain II, domain III and domain IV.
- a truncated domain IV EGFR refers to a EGFR peptide, which includes domain IV, but does not incude domain I, domain II or domain II EGFR.
- an anti-epidermal growth factor receptor (EGFR) antibody capapble of binding a truncated domain IV EGFR and not substantially binding to EGFR comprising domain I, domain II, domain II and domain IV.
- EGFR epidermal growth factor receptor
- an anti-epidermal growth factor receptor (EGFR) antibody including a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes any one of the combinations of a CDR1 sequence, a CDR2 sequence and a CDR3 sequence set forth by Table 1, 3, 5 or 7; and wherein the light chain variable domain includes any one of the combinations of a CDR1 sequence, a CDR2 sequence and a CDR3 sequence set forth by Table 2, 4, 6 or 8.
- EGFR anti-epidermal growth factor receptor
- an anti-epidermal growth factor receptor (EGFR) antibody including a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 16, a CDR H2 as set forth in SEQ ID NO: 17 and a CDR H3 as set forth in SEQ ID NO: 18; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO:52, a CDR L2 as set forth in SEQ ID NO:53, and a CDR L3 as set forth in SEQ ID NO:54.
- EGFR anti-epidermal growth factor receptor
- an anti-epidermal growth factor receptor (EGFR) antibody including a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 19, a CDR H2 as set forth in SEQ ID NO:20 and a CDR H3 as set forth in SEQ ID NO:21; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO:55, a CDR L2 as set forth in SEQ ID NO:56, and a CDR L3 as set forth in SEQ ID NO:57.
- EGFR anti-epidermal growth factor receptor
- an anti-epidermal growth factor receptor (EGFR) antibody including a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO:22, a CDR H2 as set forth in SEQ ID NO:23 and a CDR H3 as set forth in SEQ ID NO:24; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO:58, a CDR L2 as set forth in SEQ ID NO:59, and a CDR L3 as set forth in SEQ ID NO:60.
- EGFR anti-epidermal growth factor receptor
- an anti-epidermal growth factor receptor (EGFR) antibody including a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO:25, a CDR H2 as set forth in SEQ ID NO:26 and a CDR H3 as set forth in SEQ ID NO:27; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO:61, a CDR L2 as set forth in SEQ ID NO:62, and a CDR L3 as set forth in SEQ ID NO:63.
- EGFR anti-epidermal growth factor receptor
- an anti-epidermal growth factor receptor (EGFR) antibody including a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO:34, a CDR H2 as set forth in SEQ ID NO:35 and a CDR H3 as set forth in SEQ ID NO:36; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO:70, a CDR L2 as set forth in SEQ ID NO:71, and a CDR L3 as set forth in SEQ ID NO:72.
- EGFR anti-epidermal growth factor receptor
- an anti-epidermal growth factor receptor (EGFR) antibody including a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 85, a CDR H2 as set forth in SEQ ID NO: 86 and a CDR H3 as set forth in SEQ ID NO: 87; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO: 118, a CDR L2 as set forth in SEQ ID NO: 119, and a CDR L3 as set forth in SEQ ID NO: 120.
- EGFR anti-epidermal growth factor receptor
- an anti-epidermal growth factor receptor (EGFR) antibody comprising a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 88, a CDR H2 as set forth in SEQ ID NO:89 and a CDR H3 as set forth in SEQ ID NO:90; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO: 121, a CDR L2 as set forth in SEQ ID NO: 122, and a CDR L3 as set forth in SEQ ID NO: 123.
- EGFR anti-epidermal growth factor receptor
- an anti-epidermal growth factor receptor (EGFR) antibody including a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO:94, a CDR H2 as set forth in SEQ ID NO:95 and a CDR H3 as set forth in SEQ ID NO:96; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO: 127, a CDR L2 as set forth in SEQ ID NO: 128, and a CDR L3 as set forth in SEQ ID NO: 129.
- EGFR anti-epidermal growth factor receptor
- an anti-epidermal growth factor receptor (EGFR) antibody including a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO:97, a CDR H2 as set forth in SEQ ID NO:98 and a CDR H3 as set forth in SEQ ID NO:99; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO: 130, a CDR L2 as set forth in SEQ ID NO: 131, and a CDR L3 as set forth in SEQ ID NO: 132.
- EGFR anti-epidermal growth factor receptor
- an anti-epidermal growth factor receptor (EGFR) antibody including a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 100, a CDR H2 as set forth in SEQ ID NO:101 and a CDRH3 as set forth in SEQ ID NO:102; and wherein the light chain variable domain includes:a CDR LI as set forth in SEQ ID NO: 133, a CDR L2 as set forth in SEQ ID NO: 134, and a CDR L3 as set forth in SEQ ID NO: 135.
- the antibody includes a heavy chain sequence of SEQ ID NO: 247 and a light chain sequence of SEQ ID NO:248.
- an anti-epidermal growth factor receptor (EGFR) antibody including a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 103, a CDR H2 as set forth in SEQ ID NO:104 and a CDRH3 as set forth in SEQ ID NO:105; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO: 136, a CDR L2 as set forth in SEQ ID NO: 137, and a CDR L3 as set forth in SEQ ID NO: 138.
- EGFR anti-epidermal growth factor receptor
- the antibody includes a heavy chain sequence of SEQ ID NO: 249 and a light chain sequence of SEQ ID NO:250.
- an anti-epidermal growth factor receptor (EGFR) antibody including a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 139, a CDR H2 as set forth in SEQ ID NO: 140 and a CDR H3 as set forth in SEQ ID NO: 141 ; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO: 169, a CDR L2 as set forth in SEQ ID NO: 170, and a CDR L3 as set forth in SEQ ID NO: 171.
- an anti-epidermal growth factor receptor (EGFR) antibody including a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 145, a CDR H2 as set forth in SEQ ID NO:146 and a CDRH3 as set forth in SEQ ID NO:147; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO: 175, a CDR L2 as set forth in SEQ ID NO: 176, and a CDR L3 as set forth in SEQ ID NO: 177.
- EGFR anti-epidermal growth factor receptor
- an anti-epidermal growth factor receptor (EGFR) antibody including a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 148, a CDR H2 as set forth in SEQ ID NO: 149 and a CDRH3 as set forth in SEQ ID NO: 150; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO: 178, a CDR L2 as set forth in SEQ ID NO: 179, and a CDR L3 as set forth in SEQ ID NO: 180.
- the antibody includes a heavy chain sequence of SEQ ID NO:257 and a light chain sequence of SEQ ID NO:258.
- an anti-epidermal growth factor receptor (EGFR) antibody including a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 154, a CDR H2 as set forth in SEQ ID NO:155 and a CDRH3 as set forth in SEQ ID NO:156; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO: 184, a CDR L2 as set forth in SEQ ID NO: 185, and a CDR L3 as set forth in SEQ ID NO: 186.
- EGFR anti-epidermal growth factor receptor
- an anti-epidermal growth factor receptor (EGFR) antibody including a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 157, a CDR H2 as set forth in SEQ ID NO:158 and a CDRH3 as set forth in SEQ ID NO:159; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO: 187, a CDR L2 as set forth in SEQ ID NO: 188, and a CDR L3 as set forth in SEQ ID NO: 189.
- EGFR anti-epidermal growth factor receptor
- an anti-epidermal growth factor receptor (EGFR) antibody including a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 163, a CDR H2 as set forth in SEQ ID NO:164 and a CDRH3 as set forth in SEQ ID NO:165; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO: 193, a CDR L2 as set forth in SEQ ID NO: 194, and a CDR L3 as set forth in SEQ ID NO: 195.
- EGFR anti-epidermal growth factor receptor
- an anti-epidermal growth factor receptor (EGFR) antibody including a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 166, a CDR H2 as set forth in SEQ ID NO:167 and a CDRH3 as set forth in SEQ ID NO:168; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO: 196, a CDR L2 as set forth in SEQ ID NO: 197, and a CDR L3 as set forth in SEQ ID NO: 198.
- EGFR anti-epidermal growth factor receptor
- an anti-epidermal growth factor receptor (EGFR) antibody including a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 199, a CDR H2 as set forth in SEQ ID NO:200 and a CDR H3 as set forth in SEQ ID NO:201; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO:202, a CDR L2 as set forth in SEQ ID NO:203, and a CDR L3 as set forth in SEQ ID NO:204.
- the antibody includes a heavy chain sequence of SEQ ID NO: 271 and a light chain sequence of SEQ ID NO:272.
- the antibody includes a heavy chain variable domain including any one of the combinations of a CDR1 sequence, a CDR2 sequence and a CDR3 sequence set forth by Table 1 and a light chain variable domain including any one of the combinations of a CDR1 sequence, a CDR2 sequence and a CDR3 sequence set forth by Table 2.
- the antibody includes a heavy chain variable domain including any one of the combinations of a CDR1 sequence, a CDR2 sequence and a CDR3 sequence set forth by Table 1 and a light chain variable domain including any one of the combinations of a CDR1 sequence, a CDR2 sequence and a CDR3 sequence set forth by Table 4.
- the antibody includes a heavy chain variable domain including any one of the combinations of a CDR1 sequence, a CDR2 sequence and a CDR3 sequence set forth by Table 1 and a light chain variable domain including any one of the combinations of a CDR1 sequence, a CDR2 sequence and a CDR3 sequence set forth by Table 6.
- the antibody includes a heavy chain variable domain including any one of the combinations of a CDR1 sequence, a CDR2 sequence and a CDR3 sequence set forth by Table 1 and a light chain variable domain including any one of the combinations of a CDR1 sequence, a CDR2 sequence and a CDR3 sequence set forth by Table 8.
- the antibody includes a heavy chain variable domain including any one of the combinations of a CDR1 sequence, a CDR2 sequence and a CDR3 sequence set forth by Table 3 and a light chain variable domain including any one of the combinations of a CDR1 sequence, a CDR2 sequence and a CDR3 sequence set forth by Table 2.
- the antibody includes a heavy chain variable domain including any one of the combinations of a CDR1 sequence, a CDR2 sequence and a CDR3 sequence set forth by Table 3 and a light chain variable domain including any one of the combinations of a CDR1 sequence, a CDR2 sequence and a CDR3 sequence set forth by Table 4.
- the antibody includes a heavy chain variable domain including any one of the combinations of a CDR1 sequence, a CDR2 sequence and a CDR3 sequence set forth by Table 3 and a light chain variable domain including any one of the combinations of a CDR1 sequence, a CDR2 sequence and a CDR3 sequence set forth by Table 6.
- the antibody includes a heavy chain variable domain including any one of the combinations of a CDR1 sequence, a CDR2 sequence and a CDR3 sequence set forth by Table 3 and a light chain variable domain including any one of the combinations of a CDR1 sequence, a CDR2 sequence and a CDR3 sequence set forth by Table 8.
- the antibody includes a heavy chain variable domain including any one of the combinations of a CDR1 sequence, a CDR2 sequence and a CDR3 sequence set forth by Table 5 and a light chain variable domain including any one of the combinations of a CDR1 sequence, a CDR2 sequence and a CDR3 sequence set forth by Table 2.
- the antibody includes a heavy chain variable domain including any one of the combinations of a CDR1 sequence, a CDR2 sequence and a CDR3 sequence set forth by Table 5 and a light chain variable domain including any one of the combinations of a CDR1 sequence, a CDR2 sequence and a CDR3 sequence set forth by Table 4.
- the antibody includes a heavy chain variable domain including any one of the combinations of a CDR1 sequence, a CDR2 sequence and a CDR3 sequence set forth by Table 5 and a light chain variable domain including any one of the combinations of a CDR1 sequence, a CDR2 sequence and a CDR3 sequence set forth by Table 6.
- the antibody includes a heavy chain variable domain including any one of the combinations of a CDR1 sequence, a CDR2 sequence and a CDR3 sequence set forth by Table 5 and a light chain variable domain including any one of the combinations of a CDR1 sequence, a CDR2 sequence and a CDR3 sequence set forth by Table 8.
- the antibody includes a heavy chain variable domain including any one of the combinations of a CDR1 sequence, a CDR2 sequence and a CDR3 sequence set forth by Table 7 and a light chain variable domain including any one of the combinations of a CDR1 sequence, a CDR2 sequence and a CDR3 sequence set forth by Table 2.
- the antibody includes a heavy chain variable domain including any one of the combinations of a CDR1 sequence, a CDR2 sequence and a CDR3 sequence set forth by Table 7 and a light chain variable domain including any one of the combinations of a CDR1 sequence, a CDR2 sequence and a CDR3 sequence set forth by Table 4.
- the antibody includes a heavy chain variable domain including any one of the combinations of a CDR1 sequence, a CDR2 sequence and a CDR3 sequence set forth by Table 7 and a light chain variable domain including any one of the combinations of a CDR1 sequence, a CDR2 sequence and a CDR3 sequence set forth by Table 6.
- the antibody includes a heavy chain variable domain including any one of the combinations of a CDR1 sequence, a CDR2 sequence and a CDR3 sequence set forth by Table 7 and a light chain variable domain including any one of the combinations of a CDR1 sequence, a CDR2 sequence and a CDR3 sequence set forth by Table 8.
- the antibody has a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 1, a CDR H2 as set forth in SEQ ID NO:2 and a CDR H3 as set forth in SEQ ID NO:3; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO:37, a CDR L2 as set forth in SEQ ID NO:38, and a CDR L3 as set forth in SEQ ID NO:39.
- the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 1, a CDR H2 as set forth in SEQ ID NO:2 and a CDR H3 as set forth in SEQ ID NO:3
- the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO:37, a CDR L2 as set forth in SEQ ID NO:38, and a CDR L3 as set forth in SEQ ID NO:39
- the antibody has a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO:4, a CDR H2 as set forth in SEQ ID NO:5 and a CDR H3 as set forth in SEQ ID NO:6; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO:40, a CDR L2 as set forth in SEQ ID NO:41, and a CDR L3 as set forth in SEQ ID NO:42.
- the antibody has a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO:7, a CDR H2 as set forth in SEQ ID NO:8 and a CDR H3 as set forth in SEQ ID NO:9; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO:43, a CDR L2 as set forth in SEQ ID NO:44, and a CDR L3 as set forth in SEQ ID NO:45.
- the antibody has a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 10, a CDR H2 as set forth in SEQ ID NO: 11 and a CDR H3 as set forth in SEQ ID NO: 12; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO: 46, a CDR L2 as set forth in SEQ ID NO: 47, and a CDR L3 as set forth in SEQ ID NO:48.
- the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 10, a CDR H2 as set forth in SEQ ID NO: 11 and a CDR H3 as set forth in SEQ ID NO: 12
- the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO: 46, a CDR L2 as set forth in SEQ ID NO: 47, and a CDR L3 as set forth in SEQ ID NO:48
- the antibody has a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 13, a CDR H2 as set forth in SEQ ID NO: 14 and a CDR H3 as set forth in SEQ ID NO: 15; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO:49, a CDR L2 as set forth in SEQ ID NO:50, and a CDR L3 as set forth in SEQ ID NO:51.
- the antibody has a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 16, a CDR H2 as set forth in SEQ ID NO: 17 and a CDR H3 as set forth in SEQ ID NO: 18; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO:52, a CDR L2 as set forth in SEQ ID NO:53, and a CDR L3 as set forth in SEQ ID NO:54.
- the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 16, a CDR H2 as set forth in SEQ ID NO: 17 and a CDR H3 as set forth in SEQ ID NO: 18
- the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO:52, a CDR L2 as set forth in SEQ ID NO:53, and a CDR L3 as set forth in SEQ ID NO:54
- the antibody has a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 19, a CDR H2 as set forth in SEQ ID NO: 20 and a CDR H3 as set forth in SEQ ID NO:21; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO:55, a CDR L2 as set forth in SEQ ID NO:56, and a CDR L3 as set forth in SEQ ID NO:57.
- the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 19, a CDR H2 as set forth in SEQ ID NO: 20 and a CDR H3 as set forth in SEQ ID NO:21
- the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO:55, a CDR L2 as set forth in SEQ ID NO:56, and a CDR L3 as set forth in SEQ ID NO:57
- the antibody has a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 22, a CDR H2 as set forth in SEQ ID NO: 23 and a CDR H3 as set forth in SEQ ID NO:24; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO:58, a CDR L2 as set forth in SEQ ID NO:59, and a CDR L3 as set forth in SEQ ID NO:60.
- the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 22, a CDR H2 as set forth in SEQ ID NO: 23 and a CDR H3 as set forth in SEQ ID NO:24
- the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO:58, a CDR L2 as set forth in SEQ ID NO:59, and a CDR L3 as set forth in SEQ ID NO:60
- the antibody has a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO:25, a CDR H2 as set forth in SEQ ID NO:26 and a CDR H3 as set forth in SEQ ID NO: 27; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO:61, a CDR L2 as set forth in SEQ ID NO:62, and a CDR L3 as set forth in SEQ ID NO:63.
- the antibody has a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO:28, a CDR H2 as set forth in SEQ ID NO:29 and a CDR H3 as set forth in SEQ ID NO:30; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO:64, a CDR L2 as set forth in SEQ ID NO:65, and a CDR L3 as set forth in SEQ ID NO:66.
- the antibody has a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO:31, a CDR H2 as set forth in SEQ ID NO:32 and a CDR H3 as set forth in SEQ ID NO: 33; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO:67, a CDR L2 as set forth in SEQ ID NO:68, and a CDR L3 as set forth in SEQ ID NO:69.
- the antibody has a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO:34, a CDR H2 as set forth in SEQ ID NO:35 and a CDR H3 as set forth in SEQ ID NO:36; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO:70, a CDR L2 as set forth in SEQ ID NO:71, and a CDR L3 as set forth in SEQ ID NO:72.
- the antibody has a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO:73, a CDR H2 as set forth in SEQ ID NO:74 and a CDR H3 as set forth in SEQ ID NO: 75; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO: 106, a CDR L2 as set forth in SEQ ID NO: 107, and a CDR L3 as set forth in SEQ ID NO: 108.
- the antibody has a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO:76, a CDR H2 as set forth in SEQ ID NO:77 and a CDR H3 as set forth in SEQ ID NO: 78; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO: 109, a CDR L2 as set forth in SEQ ID NO: 110, and a CDR L3 as set forth in SEQ ID NO: 111.
- the antibody has a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO:79, a CDR H2 as set forth in SEQ ID NO:80 and a CDR H3 as set forth in SEQ ID NO:81; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO: 112, a CDR L2 as set forth in SEQ ID NO: 113, and a CDR L3 as set forth in SEQ ID NO: 114.
- the antibody has a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 82, a CDR H2 as set forth in SEQ ID NO: 83 and a CDR H3 as set forth in SEQ ID NO: 84; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO: 115, a CDR L2 as set forth in SEQ ID NO: 116, and a CDR L3 as set forth in SEQ ID NO: 117.
- the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 82, a CDR H2 as set forth in SEQ ID NO: 83 and a CDR H3 as set forth in SEQ ID NO: 84; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO: 115, a CDR L2 as set forth in SEQ ID NO: 116, and
- the antibody has a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 85, a CDR H2 as set forth in SEQ ID NO: 86 and a CDR H3 as set forth in SEQ ID NO: 87; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO: 118, a CDR L2 as set forth in SEQ ID NO: 119, and a CDR L3 as set forth in SEQ ID NO: 120.
- the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 85, a CDR H2 as set forth in SEQ ID NO: 86 and a CDR H3 as set forth in SEQ ID NO: 87; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO: 118, a CDR L2 as set forth in SEQ ID NO: 119, and a CDR
- the antibody has a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 88, a CDR H2 as set forth in SEQ ID NO: 89 and a CDR H3 as set forth in SEQ ID NO:90; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO: 121, a CDR L2 as set forth in SEQ ID NO: 122, and a CDR L3 as set forth in SEQ ID NO: 123.
- the antibody has a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 91, a CDR H2 as set forth in SEQ ID NO: 92 and a CDR H3 as set forth in SEQ ID NO: 93; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO: 124, a CDR L2 as set forth in SEQ ID NO: 125, and a CDR L3 as set forth in SEQ ID NO: 126.
- the antibody has a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 94, a CDR H2 as set forth in SEQ ID NO: 95 and a CDR H3 as set forth in SEQ ID NO:96; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO: 127, a CDR L2 as set forth in SEQ ID NO: 128, and a CDR L3 as set forth in SEQ ID NO: 129.
- the antibody has a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO:97, a CDR H2 as set forth in SEQ ID NO:98 and a CDR H3 as set forth in SEQ ID NO:99; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO: 130, a CDR L2 as set forth in SEQ ID NO: 131, and a CDR L3 as set forth in SEQ ID NO: 132.
- the antibody has a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 100, a CDR H2 as set forth in SEQ ID NO: 101 and a CDR H3 as set forth in SEQ ID NO: 102; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO: 133, a CDR L2 as set forth in SEQ ID NO: 134, and a CDR L3 as set forth in SEQ ID NO: 135.
- the antibody has a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 103, a CDR H2 as set forth in SEQ ID NO: 104 and a CDR H3 as set forth in SEQ ID NO: 105; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO: 136, a CDR L2 as set forth in SEQ ID NO: 137, and a CDR L3 as set forth in SEQ ID NO: 138.
- the antibody has a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 139, a CDR H2 as set forth in SEQ ID NO: 140 and a CDR H3 as set forth in SEQ ID NO: 141; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO: 169, a CDR L2 as set forth in SEQ ID NO: 170, and a CDR L3 as set forth in SEQ ID NO: 171.
- the antibody has a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 142, a CDR H2 as set forth in SEQ ID NO: 143 and a CDR H3 as set forth in SEQ ID NO: 144; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO: 172, a CDR L2 as set forth in SEQ ID NO: 173, and a CDR L3 as set forth in SEQ ID NO: 174.
- the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 142, a CDR H2 as set forth in SEQ ID NO: 143 and a CDR H3 as set forth in SEQ ID NO: 144; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO: 172, a CDR L2 as set forth in SEQ ID NO: 173, and a C
- the antibody has a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 145, a CDR H2 as set forth in SEQ ID NO: 146 and a CDR H3 as set forth in SEQ ID NO: 147; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO: 175, a CDR L2 as set forth in SEQ ID NO: 176, and a CDR L3 as set forth in SEQ ID NO: 177.
- the antibody has a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 148, a CDR H2 as set forth in SEQ ID NO: 149 and a CDR H3 as set forth in SEQ ID NO: 150; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO: 178, a CDR L2 as set forth in SEQ ID NO: 179, and a CDR L3 as set forth in SEQ ID NO: 180.
- the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 148, a CDR H2 as set forth in SEQ ID NO: 149 and a CDR H3 as set forth in SEQ ID NO: 150
- the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO: 178, a CDR L2 as set forth in SEQ ID NO: 179, and a CDR L3
- the antibody has a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 151, a CDR H2 as set forth in SEQ ID NO: 152 and a CDR H3 as set forth in SEQ ID NO: 153; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO: 181, a CDR L2 as set forth in SEQ ID NO: 182, and a CDR L3 as set forth in SEQ ID NO: 183.
- the antibody has a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 154, a CDR H2 as set forth in SEQ ID NO: 155 and a CDR H3 as set forth in SEQ ID NO: 156; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO: 184, a CDR L2 as set forth in SEQ ID NO: 185, and a CDR L3 as set forth in SEQ ID NO: 186.
- the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 154, a CDR H2 as set forth in SEQ ID NO: 155 and a CDR H3 as set forth in SEQ ID NO: 156; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO: 184, a CDR L2 as set forth in SEQ ID NO: 185, and
- the antibody has a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 157, a CDR H2 as set forth in SEQ ID NO: 158 and a CDR H3 as set forth in SEQ ID NO: 159; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO: 187, a CDR L2 as set forth in SEQ ID NO: 188, and a CDR L3 as set forth in SEQ ID NO: 189.
- the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 157, a CDR H2 as set forth in SEQ ID NO: 158 and a CDR H3 as set forth in SEQ ID NO: 159; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO: 187, a CDR L2 as set forth in SEQ ID NO: 188, and
- the antibody has a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 160, a CDR H2 as set forth in SEQ ID NO: 161 and a CDR H3 as set forth in SEQ ID NO: 162; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO: 190, a CDR L2 as set forth in SEQ ID NO: 191, and a CDR L3 as set forth in SEQ ID NO: 192.
- the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 160, a CDR H2 as set forth in SEQ ID NO: 161 and a CDR H3 as set forth in SEQ ID NO: 162; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO: 190, a CDR L2 as set forth in SEQ ID NO: 191, and a CDR L
- the antibody has a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 163, a CDR H2 as set forth in SEQ ID NO: 164 and a CDR H3 as set forth in SEQ ID NO: 165; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO: 193, a CDR L2 as set forth in SEQ ID NO: 194, and a CDR L3 as set forth in SEQ ID NO: 195.
- the antibody has a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 166, a CDR H2 as set forth in SEQ ID NO: 167 and a CDR H3 as set forth in SEQ ID NO: 168; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO: 196, a CDR L2 as set forth in SEQ ID NO: 197, and a CDR L3 as set forth in SEQ ID NO: 198.
- the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 166, a CDR H2 as set forth in SEQ ID NO: 167 and a CDR H3 as set forth in SEQ ID NO: 168; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO: 196, a CDR L2 as set forth in SEQ ID NO: 197, and
- the antibody has a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain includes: a CDR HI as set forth in SEQ ID NO: 199, a CDR H2 as set forth in SEQ ID NO:200 and a CDR H3 as set forth in SEQ ID NO:201; and wherein the light chain variable domain includes: a CDR LI as set forth in SEQ ID NO:202, a CDR L2 as set forth in SEQ ID NO:203, and a CDR L3 as set forth in SEQ ID NO: 204.
- the antibody includes any one of the heavy chain sequences set forth by Table 9, 10, 11 or 12. In embodiments, the antibody includes any one of the heavy chain sequences set forth by Table 9. In embodiments, the antibody includes any one of the heavy chain sequences set forth by Table 10. In embodiments, the antibody includes any one of the heavy chain sequences set forth by Table 11. In embodiments, the antibody includes any one of the heavy chain sequences set forth by Table 12. In embodiments, the antibody includes any one of the light chain sequences set forth by Table 9, 10, 11 or 12. In embodiments, the antibody includes any one of the light chain sequences set forth by Table 9. In embodiments, the antibody includes any one of the light chain sequences set forth by Table 10. In embodiments, the antibody includes any one of the light chain sequences set forth by Table 11. In embodiments, the antibody includes any one of the light chain sequences set forth by Table 12.
- the antibody includes any one of the heavy chain sequences set forth by Table 9 and any one of the light chain sequences set forth in Table 9. In embodiments, the antibody includes any one of the heavy chain sequences set forth by Table 9 and any one of the light chain sequences set forth in Table 10. In embodiments, the antibody includes any one of the heavy chain sequences set forth by Table 9 and any one of the light chain sequences set forth in Table 11. In embodiments, the antibody includes any one of the heavy chain sequences set forth by Table 9 and any one of the light chain sequences set forth in Table 12. In embodiments, the antibody includes any one of the heavy chain sequences set forth by Table 10 and any one of the light chain sequences set forth in Table 9. In embodiments, the antibody includes any one of the heavy chain sequences set forth by Table
- the antibody includes any one of the heavy chain sequences set forth by Table 10 and any one of the light chain sequences set forth in Table 11. In embodiments, the antibody includes any one of the heavy chain sequences set forth by Table 10 and any one of the light chain sequences set forth in Table 12. In embodiments, the antibody includes any one of the heavy chain sequences set forth by Table 11 and any one of the light chain sequences set forth in Table 9. In embodiments, the antibody includes any one of the heavy chain sequences set forth by Table 11 and any one of the light chain sequences set forth in Table 10. In embodiments, the antibody includes any one of the heavy chain sequences set forth by Table
- the antibody includes any one of the heavy chain sequences set forth by Table 11 and any one of the light chain sequences set forth in Table 12. In embodiments, the antibody includes any one of the heavy chain sequences set forth by Table 12 and any one of the light chain sequences set forth in Table 9. In embodiments, the antibody includes any one of the heavy chain sequences set forth by Table 12 and any one of the light chain sequences set forth in Table 10. In embodiments, the antibody includes any one of the heavy chain sequences set forth by Table 12 and any one of the light chain sequences set forth in Table 11. In embodiments, the antibody includes any one of the heavy chain sequences set forth by Table
- the antibody includes any one of the heavy chain sequence and light chain sequence combinations set forth by Table 9, 10, 11 or 12.
- the antibody has a heavy chain sequence of SEQ ID NO:205 and a light sequence of SEQ ID NO:206.
- the antibody has a heavy chain sequence of SEQ ID NO:207 and a light sequence of SEQ ID NO:208.
- the antibody has a heavy chain sequence of SEQ ID NO:209 and a light sequence of SEQ ID NO:210.
- the antibody has a heavy chain sequence of SEQ ID NO:211 and a light sequence of SEQ ID NO:212.
- the antibody has a heavy chain sequence of SEQ ID NO:213 and a light sequence of SEQ ID NO:214. In embodiments, the antibody has a heavy chain sequence of SEQ ID NO:215 and a light sequence of SEQ ID NO:216. In embodiments, the antibody has a heavy chain sequence of SEQ ID NO:217 and a light sequence of SEQ ID NO:218. In embodiments, the antibody has a heavy chain sequence of SEQ ID NO:219 and a light sequence of SEQ ID NO:220. In embodiments, the antibody has a heavy chain sequence of SEQ ID NO:221 and a light sequence of SEQ ID NO:222.
- the antibody has a heavy chain sequence of SEQ ID NO:223 and a light sequence of SEQ ID NO:224. In embodiments, the antibody has a heavy chain sequence of SEQ ID NO:225 and a light sequence of SEQ ID NO:226. In embodiments, the antibody has a heavy chain sequence of SEQ ID NO:227 and a light sequence of SEQ ID NO:228. In embodiments, the antibody has a heavy chain sequence of SEQ ID NO:229 and a light sequence of SEQ ID NO:230. In embodiments, the antibody has a heavy chain sequence of SEQ ID NO:231 and a light sequence of SEQ ID NO:232.
- the antibody has a heavy chain sequence of SEQ ID NO:233 and a light sequence of SEQ ID NO:234. In embodiments, the antibody has a heavy chain sequence of SEQ ID NO:235 and a light sequence of SEQ ID NO:236. In embodiments, the antibody has a heavy chain sequence of SEQ ID NO:237 and a light sequence of SEQ ID NO:238. In embodiments, the antibody has a heavy chain sequence of SEQ ID NO:239 and a light sequence of SEQ ID NO:240. In embodiments, the antibody has a heavy chain sequence of SEQ ID NO:241 and a light sequence of SEQ ID NO:242.
- the antibody has a heavy chain sequence of SEQ ID NO:243 and a light sequence of SEQ ID NO:244. In embodiments, the antibody has a heavy chain sequence of SEQ ID NO:245 and a light sequence of SEQ ID NO:246. In embodiments, the antibody has a heavy chain sequence of SEQ ID NO:247 and a light sequence of SEQ ID NO:248. In embodiments, the antibody has a heavy chain sequence of SEQ ID NO:249 and a light sequence of SEQ ID NO:250. In embodiments, the antibody has a heavy chain sequence of SEQ ID NO:251 and a light sequence of SEQ ID NO:252.
- the antibody has a heavy chain sequence of SEQ ID NO:253 and a light sequence of SEQ ID NO:254. In embodiments, the antibody has a heavy chain sequence of SEQ ID NO:255 and a light sequence of SEQ ID NO:256. In embodiments, the antibody has a heavy chain sequence of SEQ ID NO:257 and a light sequence of SEQ ID NO:258. In embodiments, the antibody has a heavy chain sequence of SEQ ID NO:259 and a light sequence of SEQ ID NO:260. In embodiments, the antibody has a heavy chain sequence of SEQ ID NO:261 and a light sequence of SEQ ID NO:262.
- the antibody has a heavy chain sequence of SEQ ID NO:263 and a light sequence of SEQ ID NO:264. In embodiments, the antibody has a heavy chain sequence of SEQ ID NO:265 and a light sequence of SEQ ID NO:266. In embodiments, the antibody has a heavy chain sequence of SEQ ID NO:267 and a light sequence of SEQ ID NO:268. In embodiments, the antibody has a heavy chain sequence of SEQ ID NO:269 and a light sequence of SEQ ID NO:270. In embodiments, the antibody has a heavy chain sequence of SEQ ID NO:271 and a light sequence of SEQ ID NO:272.
- the antibody is capable of binding to EGFR. In embodiments, the antibody is capable of binding domain IV of EGFR. In embodiments, the antibody does not substantially bind to domain I, domain II or domain III of EGFR. In embodiments, the antibody does not substantially bind to domain I of EGFR. In embodiments, the antibody does not substantially bind to domain II of EGFR. In embodiments, the antibody does not substantially bind to domain III of EGFR.
- an antibody does not substantially bind to a domain of EGFR wherein using conventional methods and compositions well known and used in the art to detect the interaction of an antibody to an epitope (e.g., immunofluorescence, Western Blot analysis, FACS analysis) do not reveal a detectable level of binding relative to a standard control (e.g., an antibody known in the art to bind to domain III of EGFR).
- a standard control e.g., an antibody known in the art to bind to domain III of EGFR.
- the antibody binds a truncated domain IV EGFR and does not substantially bind to EGFR comprising domain I, domain II, domain II and domain IV.
- the antibody provided herein including embodiments thereof may be a humanized antibody, a Fab' fragment, a single chain antibody (scFv) or a chimeric antibody.
- the antibody is a humanized antibody.
- antibody is a Fab' fragment.
- the antibody is a scFv.
- the antibody is a chimeric antibody.
- the antibody includes a fragment crystallizable (Fc) domain.
- the Fc domain binds an effector cell ligand.
- effector cell ligand refers to a cell surface molecule expressed on an effector cell of the immune system (e.g., a cytotoxic T cell, a helper T cell, a B cell, a natural killer cell).
- the effector cell Upon binding of the antibody to the effector cell ligand expressed on the effector cell, the effector cell is activated and able to exert its function (e.g., selective killing or eradication of malignant, infected or otherwise unhealthy cells).
- the effector cell ligand is a CD3 protein.
- the effector cell ligand is a CD 16 protein.
- the CD 16 protein includes a valine at a position corresponding to the position of amino acid residue 158.
- the CD16 protein includes a phenylalanine at a position corresponding to the position of amino acid residue 158.
- the effector cell ligand is a CD32 protein.
- the effector cell ligand is a NKp46 protein.
- the Fc domain includes an effector cell inhibiting substitution. In the presence of an effector cell inhibiting substitution the binding of the Fc domain to the effector cell ligand decreases activation of an effector cell relative to the absence of said substitution.
- the binding of the Fc domain to the effector cell ligand results in substantially no activation of an effector cell relative to the absence of said substitution.
- the Fc domain includes a N297G substitution, a R292C substitution, a V302C substitution or a combination thereof.
- the Fc domain includes a N297G substitution.
- the Fc domain includes a R292C substitution.
- the Fc domain includes a V302C substitution.
- the effector cell inhibiting substitution is aN297G substitution, a R292C substitution, or a V302C substitution.
- the Fc domain includes an effector cell enhancing substitution. In the presence of an effector cell enhancing substitution the binding of the Fc domain to the effector cell ligand increases activation of an effector cell relative to the absence of said substitution.
- the Fc domain includes a S239D substitution, a I332E substitution or a combination thereof.
- the Fc domain includes a S239D substitution.
- the Fc domain includes a I332E substitution.
- the effector cell enhancing substitution is a S239D substitution, or a I332E substitution.
- the antibody provided herein is an IgG.
- the antibody is a human IgGi.
- the antibody is capable of eliciting antibody- dependent cell mediated cytotoxicity (ADCC).
- the antibody is a human IgG2.
- the antibody is a human IgG2 and the antibody does not elicit ADCC. Wherein the antibody does not elicit ADCC, ADCC is not elicited in a detectable amount. In embodiments, the antibody does not elicit target cell killing in the presence of an effector cell at a detectable amount. Standard methods and compositions conventional in the biological arts are contemplated for detecting ADCC using the antibodies provided herein.
- the Fab’ domain of an antibody provided herein including embodiments thereof may bind its epitope (i.e. EGFR) with a binding affinity that is different relative to the binding affinity of the IgG isotype of that same antibody.
- the Fab domain of the anti-EGFR antibody binds EGFR with a greater KD relative to said IgG.
- a monovalent form of the antibody binds EGFR with a greater equilibrium dissociation constant (KD) relative to a bivalent form of the antibody.
- KD equilibrium dissociation constant
- the monovalent form binds EGFR with about 100- to 1000-fold greater KD relative to said bivalent form.
- the monovalent form binds EGFR with about 200- to 400-fold greater KD relative to said bivalent form.
- the Fab’ fragment binds EGFR with a greater equilibrium dissociation constant (KD) relative to the IgG (e.g., human IgGi or human IgG2). In embodiments, the Fab’ fragment binds EGFR with about 100- to 1000-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 150- to 1000- fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 200- to 1000-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 250- to 1000-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 300- to 1000-fold greater KD relative to the IgG.
- KD equilibrium dissociation constant
- the Fab’ fragment binds EGFR with about 350- to 1000-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 400- to 1000- fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 450- to 1000-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 500- to 1000-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 550- to 1000-fold greater KD relative to the IgG.
- the Fab’ fragment binds EGFR with about 600- to 1000-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 650- to 1000- fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 700- to 1000-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 750- to 1000-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 800- to 1000-fold greater KD relative to the IgG.
- the Fab’ fragment binds EGFR with about 850- to 1000-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 900- to 1000- fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 950- to 1000-fold greater KD relative to the IgG.
- the Fab’ fragment binds EGFR with about 100- to 950-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 100- to 900-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 100- to 850-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 100- to 800-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 100- to 750-fold greater KD relative to the IgG.
- the Fab’ fragment binds EGFR with about 100- to 700-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 100- to 650-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 100- to 600-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 100- to 550-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 100- to 500-fold greater KD relative to the IgG.
- the Fab’ fragment binds EGFR with about 100- to 450-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 100- to 400-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 100- to 350-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 100- to 300-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 100- to 350-fold greater KD relative to the IgG.
- the Fab’ fragment binds EGFR with about 100- to 200-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 100- to 150-fold greater KD relative to the IgG.
- the Fab’ fragment binds EGFR with about 100-, 150-, 200-, 250-, 300-, 350-, 400-, 450-, 500-, 550-, 600-, 650-, 700-, 750-, 800-, 850-, 900-, 950-, or 1000- fold greater KD relative to the IgG.
- the Fab’ fragment binds EGFR with 100- to 1000-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 150- to 1000-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 200- to 1000-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 250- to 1000-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 300- to 1000-fold greater KD relative to the IgG.
- the Fab’ fragment binds EGFR with 350- to 1000-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 400- to 1000-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 450- to 1000-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 500- to 1000-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 550- to 1000-fold greater KD relative to the IgG.
- the Fab’ fragment binds EGFR with 600- to 1000-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 650- to 1000-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 700- to 1000-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 750- to 1000-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 800- to 1000-fold greater KD relative to the IgG.
- the Fab’ fragment binds EGFR with 850- to 1000-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 900- to 1000-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 950- to 1000-fold greater KD relative to the IgG.
- the Fab’ fragment binds EGFR with 100- to 950-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 100- to 900-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 100- to 850-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 100- to 800-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 100- to 750-fold greater KD relative to the IgG.
- the Fab’ fragment binds EGFR with 100- to 700-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 100- to 650-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 100- to 600-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 100- to 550-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 100- to 500-fold greater KD relative to the IgG.
- the Fab’ fragment binds EGFR with 100- to 450-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 100- to 400-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 100- to 350-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 100- to 300-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 100- to 350-fold greater KD relative to the IgG.
- the Fab’ fragment binds EGFR with 100- to 200-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 100- to 150-fold greater KD relative to the IgG.
- the Fab’ fragment binds EGFR with 100-, 150-, 200-, 250-, 300-, 350-, 400-, 450-, 500-, 550-, 600-, 650-, 700-, 750-, 800-, 850-, 900-, 950-, or 1000-fold greater KD relative to the IgG.
- the Fab’ fragment binds EGFR with about 200- to 400-fold greater KD relative to the IgG.
- the Fab’ fragment binds EGFR with about 210- to 400-fold greater KD relative to the IgG.
- the Fab’ fragment binds EGFR with about 220- to 400-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 230- to 400-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 240- to 400-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 250- to 400-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 260- to 400-fold greater KD relative to the IgG.
- the Fab’ fragment binds EGFR with about 270- to 400-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 280- to 400-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 290- to 400-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 300- to 400-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 310- to 400-fold greater KD relative to the IgG.
- the Fab’ fragment binds EGFR with about 320- to 400-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 330- to 400-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 340- to 400-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 350- to 400-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 360- to 400-fold greater KD relative to the IgG.
- the Fab’ fragment binds EGFR with about 370- to 400-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 380- to 400-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 390- to 400-fold greater KD relative to the IgG.
- the Fab’ fragment binds EGFR with about 200- to 390-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 200- to 380-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 200- to 370-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 200- to 360-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 200- to 350-fold greater KD relative to the IgG.
- the Fab’ fragment binds EGFR with about 200- to 340-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 200- to 330-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 200- to 320-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 200- to 310-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 200- to 300-fold greater KD relative to the IgG.
- the Fab’ fragment binds EGFR with about 200- to 290-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 200- to 280-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 200- to 270-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 200- to 260-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 200- to 250-fold greater KD relative to the IgG.
- the Fab’ fragment binds EGFR with about 200- to 240-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 200- to 230-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 200- to 220-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with about 200- to 210-fold greater KD relative to the IgG.
- the Fab’ fragment binds EGFR with about 200-, 210-, 220-, 230-, 240-, 250-, 260-, 270-, 280-, 290-, 300-, 310-, 320-, 330-, 340-, 340-, 350-, 370-, 380-, 390- or 400-fold greater KD relative to the IgG.
- the Fab’ fragment binds EGFR with 200- to 400-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 210- to 400-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 220- to 400-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 230- to 400-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 240- to 400-fold greater KD relative to the IgG.
- the Fab’ fragment binds EGFR with 250- to 400-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 260- to 400-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 270- to 400-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 280- to 400-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 290- to 400-fold greater KD relative to the IgG.
- the Fab’ fragment binds EGFR with 300- to 400-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 310- to 400-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 320- to 400-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 330- to 400-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 340- to 400-fold greater KD relative to the IgG.
- the Fab’ fragment binds EGFR with 350- to 400-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 360- to 400-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 370- to 400-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 380- to 400-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 390- to 400-fold greater KD relative to the IgG.
- the Fab’ fragment binds EGFR with 200- to 390-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 200- to 380-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 200- to 370-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 200- to 360-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 200- to 350-fold greater KD relative to the IgG.
- the Fab’ fragment binds EGFR with 200- to 340-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 200- to 330-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 200- to 320-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 200- to 310-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 200- to 300-fold greater KD relative to the IgG.
- the Fab’ fragment binds EGFR with 200- to 290-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 200- to 280-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 200- to 270-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 200- to 260-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 200- to 250-fold greater KD relative to the IgG.
- the Fab’ fragment binds EGFR with 200- to 240-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 200- to 230-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 200- to 220-fold greater KD relative to the IgG. In embodiments, the Fab’ fragment binds EGFR with 200- to 210-fold greater KD relative to the IgG.
- the Fab’ fragment binds EGFR with 200-, 210-, 220-, 230-, 240-, 250-, 260-, 270-, 280-, 290-, 300-, 310-, 320-, 330-, 340-, 340-, 350-, 370-, 380-, 390- or 400-fold greater KD relative to the IgG.
- the Fab’ fragment binds EGFR with a KD of about 100 nM to about 500 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of about 120 nM to about 500 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of about 140 nM to about 500 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of about 160 nM to about 500 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of about 180 nM to about 500 nM.
- the Fab’ fragment binds EGFR with a KD of about 200 nM to about 500 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of about 220 nM to about 500 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of about 240 nM to about 500 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of about 260 nM to about 500 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of about 280 nM to about 500 nM.
- the Fab’ fragment binds EGFR with a KD of about 300 nM to about 500 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of about 320 nM to about 500 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of about 340 nM to about 500 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of about 360 nM to about 500 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of about 380 nM to about 500 nM.
- the Fab’ fragment binds EGFR with a KD of about 400 nM to about 500 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of about 420 nM to about 500 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of about 440 nM to about 500 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of about 460 nM to about 500 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of about 480 nM to about 500 nM.
- the Fab’ fragment binds EGFR with a KD of about 100 nM to about 480 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of about 100 nM to about 460 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of about 100 nM to about 440 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of about 100 nM to about 420 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of about 100 nM to about 400 nM.
- the Fab’ fragment binds EGFR with a KD of about 100 nM to about 380 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of about 100 nM to about 360 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of about 100 nM to about 340 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of about 100 nM to about 320 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of about 100 nM to about 300 nM.
- the Fab’ fragment binds EGFR with a KD of about 100 nM to about 280 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of about 100 nM to about 260 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of about 100 nM to about 240 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of about 100 nM to about 220 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of about 100 nM to about 200 nM.
- the Fab’ fragment binds EGFR with a KD of about 100 nM to about 180 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of about 100 nM to about 160 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of about 100 nM to about 140 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of about 100 nM to about 120 nM.
- the Fab’ fragment binds EGFR with a KD of about 100 nM, 120 nM, 140 nM, 160 nM, 180 nM, 200 nM, 220 nM, 240 nM, 260 nM, 280 nM, 300 nM, 320 nM, 340 nM, 360 nM, 380 nM, 400 nM, 420 nM, 440 nM, 460 nM, 480 nM, or 500 nM.
- the Fab’ fragment binds EGFR with a KD of 100 nM to 500 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of 120 nM to 500 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of 140 nM to 500 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of 160 nM to 500 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of 180 nM to 500 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of 200 nM to 500 nM.
- the Fab’ fragment binds EGFR with a KD of 220 nM to 500 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of 240 nM to 500 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of 260 nM to 500 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of 280 nM to 500 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of 300 nM to 500 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of 320 nM to 500 nM.
- the Fab’ fragment binds EGFR with a KD of 340 nM to 500 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of 360 nM to 500 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of 380 nM to 500 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of 400 nM to 500 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of 420 nM to 500 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of 440 nM to 500 nM.
- the Fab’ fragment binds EGFR with a KD of 460 nM to 500 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of 480 nM to 500 nM.
- the Fab’ fragment binds EGFR with a KD of 100 nM to 480 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of 100 nM to 460 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of 100 nM to 440 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of 100 nM to 420 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of 100 nM to 400 nM.
- the Fab’ fragment binds EGFR with a KD of 100 nM to 380 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of 100 nM to 360 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of 100 nM to 340 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of 100 nM to 320 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of 100 nM to 300 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of 100 nM to 280 nM.
- the Fab’ fragment binds EGFR with a KD of 100 nM to 260 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of 100 nM to 240 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of 100 nM to 220 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of 100 nM to 200 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of 100 nM to 180 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of 100 nM to 160 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of 100 nM to 140 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of 100 nM to 120 nM.
- the Fab’ fragment binds EGFR with a K D of 100 nM, 120 nM, 140 nM, 160 nM, 180 nM, 200 nM, 220 nM, 240 nM, 260 nM, 280 nM, 300 nM, 320 nM, 340 nM, 360 nM, 380 nM, 400 nM, 420 nM, 440 nM, 460 nM, 480 nM, or 500 nM.
- the Fab’ fragment binds EGFR with a KD of about 170 nM. In embodiments, the Fab’ fragment binds EGFR with a KD of 170 nM.
- the IgG binds EGFR with a K D from about 100 pM to 1000 pM. In embodiments, the IgG binds EGFR with a K D from about 120 pM to 1000 pM. In embodiments, the IgG binds EGFR with a K D from about 140 pM to 1000 pM. In embodiments, the IgG binds EGFR with a KD from about 160 pM to 1000 pM. In embodiments, the IgG binds EGFR with a K D from about 180 pM to 1000 pM. In embodiments, the IgG binds EGFR with a KD from about 200 pM to 1000 pM.
- the IgG binds EGFR with a KD from about 220 pM to 1000 pM. In embodiments, the IgG binds EGFR with a K D from about 240 pM to 1000 pM. In embodiments, the IgG binds EGFR with a KD from about 260 pM to 1000 pM. In embodiments, the IgG binds EGFR with a K D from about 280 pM to 1000 pM. In embodiments, the IgG binds EGFR with a K D from about 300 pM to 1000 pM. In embodiments, the IgG binds EGFR with a K D from about 320 pM to 1000 pM.
- the IgG binds EGFR with a K D from about 340 pM to 1000 pM. In embodiments, the IgG binds EGFR with a K D from about 360 pM to 1000 pM. In embodiments, the IgG binds EGFR with a K D from about 380 pM to 1000 pM. In embodiments, the IgG binds EGFR with a K D from about 400 pM to 1000 pM. In embodiments, the IgG binds EGFR with a K D from about 420 pM to 1000 pM. In embodiments, the IgG binds EGFR with a KD from about 440 pM to 1000 pM.
- the IgG binds EGFR with a K D from about 460 pM to 1000 pM. In embodiments, the IgG binds EGFR with a KD from about 480 pM to 1000 pM. In embodiments, the IgG binds EGFR with a KD from about 500 pM to 1000 pM. In embodiments, the IgG binds EGFR with a K D from about 520 pM to 1000 pM. In embodiments, the IgG binds EGFR with a KD from about 540 pM to 1000 pM. In embodiments, the IgG binds EGFR with a K D from about 560 pM to 1000 pM.
- the IgG binds EGFR with a K D from about 580 pM to 1000 pM. In embodiments, the IgG binds EGFR with a K D from about 600 pM to 1000 pM. In embodiments, the IgG binds EGFR with a KD from about 620 pM to 1000 pM. In embodiments, the IgG binds EGFR with a K D from about 640 pM to 1000 pM. In embodiments, the IgG binds EGFR with a KD from about 660 pM to 1000 pM.
- the IgG binds EGFR with a KD from about 680 pM to 1000 pM. In embodiments, the IgG binds EGFR with a K D from about 700 pM to 1000 pM. In embodiments, the IgG binds EGFR with a KD from about 720 pM to 1000 pM. In embodiments, the IgG binds EGFR with a K D from about 740 pM to 1000 pM. In embodiments, the IgG binds EGFR with a K D from about 760 pM to 1000 pM. In embodiments, the IgG binds EGFR with a K D from about 780 pM to 1000 pM.
- the IgG binds EGFR with a K D from about 800 pM to 1000 pM. In embodiments, the IgG binds EGFR with a KD from about 820 pM to 1000 pM. In embodiments, the IgG binds EGFR with a K D from about 840 pM to 1000 pM. In embodiments, the IgG binds EGFR with a KD from about 860 pM to 1000 pM. In embodiments, the IgG binds EGFR with a KD from about 880 pM to 1000 pM. In embodiments, the IgG binds EGFR with a K D from about 900 pM to 1000 pM.
- the IgG binds EGFR with a KD from about 920 pM to 1000 pM. In embodiments, the IgG binds EGFR with a K D from about 940 pM to 1000 pM. In embodiments, the IgG binds EGFR with a K D from about 960 pM to 1000 pM. In embodiments, the IgG binds EGFR with a K D from about 980 pM to 1000 pM.
- the IgG binds EGFR with a K D from about 100 pM to 980 pM. In embodiments, the IgG binds EGFR with a K D from about 100 pM to 960 pM. In embodiments, the IgG binds EGFR with a KD from about 100 pM to 940 pM. In embodiments, the IgG binds EGFR with a K D from about 100 pM to 920 pM. In embodiments, the IgG binds EGFR with a K D from about 100 pM to 900 pM.
- the IgG binds EGFR with a KD from about 100 pM to 880 pM. In embodiments, the IgG binds EGFR with a K D from about 100 pM to 860 pM. In embodiments, the IgG binds EGFR with a K D from about 100 pM to 840 pM. In embodiments, the IgG binds EGFR with a K D from about 100 pM to 820 pM. In embodiments, the IgG binds EGFR with a K D from about 100 pM to 800 pM. In embodiments, the IgG binds EGFR with a K D from about 100 pM to 780 pM.
- the IgG binds EGFR with a K D from about 100 pM to 760 pM. In embodiments, the IgG binds EGFR with a K D from about 100 pM to 740 pM. In embodiments, the IgG binds EGFR with a K D from about 100 pM to 720 pM. In embodiments, the IgG binds EGFR with a KD from about 100 pM to 700 pM. In embodiments, the IgG binds EGFR with a K D from about 100 pM to 680 pM. In embodiments, the IgG binds EGFR with a KD from about 100 pM to 660 pM.
- the IgG binds EGFR with a KD from about 100 pM to 640 pM. In embodiments, the IgG binds EGFR with a K D from about 100 pM to 620 pM. In embodiments, the IgG binds EGFR with a KD from about 100 pM to 600 pM. In embodiments, the IgG binds EGFR with a K D from about 100 pM to 580 pM. In embodiments, the IgG binds EGFR with a K D from about 100 pM to 560 pM.
- the IgG binds EGFR with a KD from about 100 pM to 540 pM. In embodiments, the IgG binds EGFR with a K D from about 100 pM to 520 pM. In embodiments, the IgG binds EGFR with a KD from about 100 pM to 500 pM. In embodiments, the IgG binds EGFR with a KD from about 100 pM to 480 pM. In embodiments, the IgG binds EGFR with a K D from about 100 pM to 460 pM.
- the IgG binds EGFR with a KD from about 100 pM to 440 pM. In embodiments, the IgG binds EGFR with a K D from about 100 pM to 420 pM. In embodiments, the IgG binds EGFR with a K D from about 100 pM to 400 pM. In embodiments, the IgG binds EGFR with a K D from about 100 pM to 380 pM. In embodiments, the IgG binds EGFR with a K D from about 100 pM to 360 pM. In embodiments, the IgG binds EGFR with a K D from about 100 pM to 340 pM.
- the IgG binds EGFR with a K D from about 100 pM to 320 pM. In embodiments, the IgG binds EGFR with a K D from about 100 pM to 300 pM. In embodiments, the IgG binds EGFR with a K D from about 100 pM to 280 pM. In embodiments, the IgG binds EGFR with a KD from about 100 pM to 260 pM. In embodiments, the IgG binds EGFR with a K D from about 100 pM to 240 pM. In embodiments, the IgG binds EGFR with a KD from about 100 pM to 220 pM.
- the IgG binds EGFR with a KD from about 100 pM to 200 pM. In embodiments, the IgG binds EGFR with a K D from about 100 pM to 180 pM. In embodiments, the IgG binds EGFR with a KD from about 100 pM to 160 pM. In embodiments, the IgG binds EGFR with a K D from about 100 pM to 140 pM. In embodiments, the IgG binds EGFR with a K D from about 100 pM to 120 pM.
- the IgG binds EGFR with a K D from 100 pM to 1000 pM. In embodiments, the IgG binds EGFR with a KD from 120 pM to 1000 pM. In embodiments, the IgG binds EGFR with a K D from 140 pM to 1000 pM. In embodiments, the IgG binds EGFR with a KD from 160 pM to 1000 pM. In embodiments, the IgG binds EGFR with a KD from 180 pM to 1000 pM. In embodiments, the IgG binds EGFR with a KD from 200 pM to 1000 pM.
- the IgG binds EGFR with a K D from 220 pM to 1000 pM. In embodiments, the IgG binds EGFR with a KD from 240 pM to 1000 pM. In embodiments, the IgG binds EGFR with a K D from 260 pM to 1000 pM. In embodiments, the IgG binds EGFR with a K D from 280 pM to 1000 pM. In embodiments, the IgG binds EGFR with a K D from 300 pM to 1000 pM. In embodiments, the IgG binds EGFR with a K D from 320 pM to 1000 pM.
- the IgG binds EGFR with a K D from 340 pM to 1000 pM. In embodiments, the IgG binds EGFR with a KD from 360 pM to 1000 pM. In embodiments, the IgG binds EGFR with a KD from 380 pM to 1000 pM. In embodiments, the IgG binds EGFR with a KD from 400 pM to 1000 pM. In embodiments, the IgG binds EGFR with a KD from 420 pM to 1000 pM. In embodiments, the IgG binds EGFR with a KD from 440 pM to 1000 pM.
- the IgG binds EGFR with a KD from 460 pM to 1000 pM. In embodiments, the IgG binds EGFR with a KD from 480 pM to 1000 pM. In embodiments, the IgG binds EGFR with a KD from 500 pM to 1000 pM.
- the IgG binds EGFR with a K D from 520 pM to 1000 pM. In embodiments, the IgG binds EGFR with a K D from 540 pM to 1000 pM. In embodiments, the IgG binds EGFR with a K D from 560 pM to 1000 pM. In embodiments, the IgG binds EGFR with a K D from 580 pM to 1000 pM. In embodiments, the IgG binds EGFR with a K D from 600 pM to 1000 pM. In embodiments, the IgG binds EGFR with a KD from 620 pM to 1000 pM.
- the IgG binds EGFR with a K D from 640 pM to 1000 pM. In embodiments, the IgG binds EGFR with a K D from 660 pM to 1000 pM. In embodiments, the IgG binds EGFR with a KD from 680 pM to 1000 pM. In embodiments, the IgG binds EGFR with a K D from 700 pM to 1000 pM. In embodiments, the IgG binds EGFR with a K D from 720 pM to 1000 pM. In embodiments, the IgG binds EGFR with a K D from 740 pM to 1000 pM.
- the IgG binds EGFR with a K D from 760 pM to 1000 pM. In embodiments, the IgG binds EGFR with a K D from 780 pM to 1000 pM. In embodiments, the IgG binds EGFR with a K D from 800 pM to 1000 pM. In embodiments, the IgG binds EGFR with a K D from 820 pM to 1000 pM. In embodiments, the IgG binds EGFR with a K D from 840 pM to 1000 pM. In embodiments, the IgG binds EGFR with a K D from 860 pM to 1000 pM.
- the IgG binds EGFR with a K D from 880 pM to 1000 pM. In embodiments, the IgG binds EGFR with a KD from 900 pM to 1000 pM. In embodiments, the IgG binds EGFR with a K D from 920 pM to 1000 pM. In embodiments, the IgG binds EGFR with a KD from 940 pM to 1000 pM. In embodiments, the IgG binds EGFR with a KD from 960 pM to 1000 pM. In embodiments, the IgG binds EGFR with a KD from 980 pM to 1000 pM.
- the IgG binds EGFR with a K D from 100 pM to 980 pM. In embodiments, the IgG binds EGFR with a K D from 100 pM to 960 pM. In embodiments, the IgG binds EGFR with a K D from 100 pM to 940 pM. In embodiments, the IgG binds EGFR with a K D from 100 pM to 920 pM. In embodiments, the IgG binds EGFR with a K D from 100 pM to 900 pM. In embodiments, the IgG binds EGFR with a KD from 100 pM to 880 pM.
- the IgG binds EGFR with a K D from 100 pM to 860 pM. In embodiments, the IgG binds EGFR with a KD from 100 pM to 840 pM. In embodiments, the IgG binds EGFR with a KD from 100 pM to 820 pM. In embodiments, the IgG binds EGFR with a K D from 100 pM to 800 pM. In embodiments, the IgG binds EGFR with a K D from 100 pM to 780 pM. In embodiments, the IgG binds EGFR with a KD from 100 pM to 760 pM.
- the IgG binds EGFR with a K D from 100 pM to 740 pM. In embodiments, the IgG binds EGFR with a K D from 100 pM to 720 pM. In embodiments, the IgG binds EGFR with a K D from 100 pM to 700 pM. In embodiments, the IgG binds EGFR with a K D from 100 pM to 680 pM. In embodiments, the IgG binds EGFR with a K D from 100 pM to 660 pM.
- the IgG binds EGFR with a KD from 100 pM to 640 pM. In embodiments, the IgG binds EGFR with a KD from 100 pM to 620 pM. In embodiments, the IgG binds EGFR with a KD from 100 pM to 600 pM. In embodiments, the IgG binds EGFR with a KD from 100 pM to 580 pM. In embodiments, the IgG binds EGFR with a KD from 100 pM to 560 pM. In embodiments, the IgG binds EGFR with a KD from 100 pM to 540 pM.
- the IgG binds EGFR with a KD from 100 pM to 520 pM. In embodiments, the IgG binds EGFR with a KD from 100 pM to 500 pM. In embodiments, the IgG binds EGFR with a KD from 100 pM to 480 pM. In embodiments, the IgG binds EGFR with a KD from 100 pM to 460 pM. In embodiments, the IgG binds EGFR with a KD from 100 pM to 440 pM. In embodiments, the IgG binds EGFR with a KD from 100 pM to 420 pM.
- the IgG binds EGFR with a KD from 100 pM to 400 pM. In embodiments, the IgG binds EGFR with a KD from 100 pM to 380 pM. In embodiments, the IgG binds EGFR with a KD from 100 pM to 360 pM. In embodiments, the IgG binds EGFR with a KD from 100 pM to 340 pM. In embodiments, the IgG binds EGFR with a KD from 100 pM to 320 pM. In embodiments, the IgG binds EGFR with a KD from 100 pM to 300 pM.
- the IgG binds EGFR with a KD from 100 pM to 280 pM. In embodiments, the IgG binds EGFR with a KD from 100 pM to 260 pM. In embodiments, the IgG binds EGFR with a KD from 100 pM to 240 pM. In embodiments, the IgG binds EGFR with a KD from 100 pM to 220 pM. In embodiments, the IgG binds EGFR with a KD from 100 pM to 200 pM. In embodiments, the IgG binds EGFR with a KD from 100 pM to 180 pM.
- the IgG binds EGFR with a KD from 100 pM to 160 pM. In embodiments, the IgG binds EGFR with a KD from 100 pM to 140 pM. In embodiments, the IgG binds EGFR with a K D from 100 pM to 120 pM.
- the IgG binds EGFR with a KD of about 100 pM, 120 pM, 140 pM, 160 pM, 180 pM, 200 pM, 220 pM, 240 pM, 260 pM, 280 pM, 300 pM, 320 pM, 340 pM,
- the IgG binds EGFR with a KD of 100 pM, 120 pM, 140 pM, 160 pM, 180 pM, 200 pM, 220 pM, 240 pM, 260 pM, 280 pM, 300 pM, 320 pM,
- the IgG binds EGFR with a KD of about 487 pM. In embodiments, the IgG binds EGFR with a KD of 487 pM. In embodiments, the IgG binds EGFR with a KD of about 214 pM. In embodiments, the IgG binds EGFR with a KD of 214 pM.
- the antibodies provided herein including embodiments thereof are capable of binding to EGFR.
- the antibody binds to EGFR.
- the antibody binds to domain IV of EGFR.
- domain IV of EGFR includes the amino acid sequence of SEQ ID NO:276.
- domain IV of EGFR is the amino acid sequence of SEQ ID NO:276.
- the antibody binds to the amino acid sequence of SEQ ID NO:276.
- the antibody binds to an amino acid sequence with 75%, 80%, 85%, 90, 95%, 98% or 99% sequence identity to human domain IV EGFR, variants or homologs thereof.
- the antibody binds to an amino acid sequence with 75%, 80%, 85%, 90, 95%, 98% or 99% sequence identity to the amino acid sequence of SEQ ID NO:276.
- the variants or homologs have at least 90%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity across the whole sequence or a portion of the sequence (e.g. a 50, 100, 150 or 200 continuous amino acid portion) of domain IV EGFR.
- the antibody binds to an amino acid sequence with at least 75%, 80%, 85%, 90, 95%, 98% or 99% sequence identity to human domain IV EGFR, wherein the identity is across the whole sequence or a portion of the sequence (e.g.
- the antibody binds to an amino acid sequence with at least 75%, 80%, 85%, 90, 95%, 98% or 99% sequence identity to SEQ ID NO:276, wherein the identity is across the whole sequence or a portion of the sequence (e.g. a 50, 100, 150 or 200 continuous amino acid portion).
- the domain IV EGFR is substantially identical to the domain IV of the protein identified by the UniProt reference number P00533 or a variant or homolog having substantial identity thereto.
- the domain IV EGFR has at least 90%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity across the whole sequence or a portion of the sequence (e.g. a 50, 100, 150 or 200 continuous amino acid portion) of the sequence of SEQ ID NO:276.
- the EGFR includes the amino acid sequence of SEQ ID NO:273,
- the EGFR includes the amino acid sequence of SEQ ID NO:273. In embodiments, the EGFR includes the amino acid sequence of SEQ ID NO:274. In embodiments, the EGFR includes the amino acid sequence of SEQ ID NO:275.
- the antibody binds the amino acid sequence of SEQ ID NO:273,
- the antibody binds the amino acid sequence of SEQ ID NO:273. In embodiments, the antibody binds the amino acid sequence of SEQ ID NO:274. In embodiments, the antibody binds the amino acid sequence of SEQ ID NO:275.
- an antibody provided herein including embodiments thereof bound to domain IV of EGFR.
- an anti-EGFR antibody capable of binding the same EGFR epitope (e.g. domain IV of EGFR) that is bound by the antibodies provided herein including embodiments thereof.
- an anti-EGFR antibody wherein the anti-EGFR antibody binds the same epitope as an antibody including: a heavy chain variable domain including any one of the combinations of a CDR 1 sequence, a CDR2 sequence and a CDR3 sequence set forth by Table 1, 3, 5 or 7; and a light chain variable domain comprising any one of the combinations of a CDR 1 sequence, a CDR2 sequence and a CDR3 sequence set forth by Table 2, 4, 6 or 8.
- the antibody may bind the same epitope as an anti-EGFR antibody including a heavy chain variable domain including: a CDR HI as set forth in SEQ ID NO: 199, a CDR H2 as set forth in SEQ ID NO: 200 and a CDR H3 as set forth in SEQ ID NO:201; and a light chain variable domain including: a CDR LI as set forth in SEQ ID NO:202, a CDR L2 as set forth in SEQ ID NO:203, and a CDR L3 as set forth in SEQ ID NO: 204.
- a heavy chain variable domain including: a CDR HI as set forth in SEQ ID NO: 199, a CDR H2 as set forth in SEQ ID NO: 200 and a CDR H3 as set forth in SEQ ID NO:201
- a light chain variable domain including: a CDR LI as set forth in SEQ ID NO:202, a CDR L2 as set forth in SEQ ID NO:203, and a CDR L3 as set forth in
- an anti-EGFR antibody wherein the anti-EGFR antibody binds the same epitope as an antibody including: a heavy chain variable domain including a CDR HI as set forth in SEQ ID NO: 199, a CDR H2 as set forth in SEQ ID NO:200 and a CDR H3 as set forth in SEQ ID NO:201; and a light chain variable domain including a CDR LI as set forth in SEQ ID NO:202, a CDR L2 as set forth in SEQ ID NO:203, and a CDR L3 as set forth in SEQ ID NO:204.
- the heavy chain sequence has the sequence of SEQ ID NO:271 and the light chain sequence has the sequence of SEQ ID NO:272.
- an anti-EGFR antibody wherein the anti-EGFR antibody binds the same epitope as an antibody including: a heavy chain variable domain including a CDR HI as set forth in SEQ ID NO: 103, a CDR H2 as set forth in SEQ ID NO: 104 and a CDR H3 as set forth in SEQ ID NO: 105; and a light chain variable domain including a CDR LI as set forth in SEQ ID NO: 136, a CDR L2 as set forth in SEQ ID NO: 137, and a CDR L3 as set forth in SEQ ID NO: 138.
- the heavy chain sequence has the sequence of SEQ ID NO:249 and said light chain sequence has the sequence of SEQ ID NO:250
- an anti-EGFR antibody wherein the anti-EGFR antibody binds the same epitope as an antibody including: a heavy chain variable domain including a CDR HI as set forth in SEQ ID NO: 148, a CDR H2 as set forth in SEQ ID NO: 149 and a CDR H3 as set forth in SEQ ID NO: 150; and a light chain variable domain including a CDR LI as set forth in SEQ ID NO: 178, a CDR L2 as set forth in SEQ ID NO: 179, and a CDR L3 as set forth in SEQ ID NO: 180.
- the heavy chain sequence has the sequence of SEQ ID NO:257 and said light chain sequence has the sequence of SEQ ID NO:258.
- an anti-EGFR antibody wherein the anti-EGFR antibody binds the same epitope as an antibody including: a heavy chain variable domain including a CDR HI as set forth in SEQ ID NO: 100, a CDR H2 as set forth in SEQ ID NO: 101 and a CDR H3 as set forth in SEQ ID NO: 102; and a light chain variable domain including a CDR LI as set forth in SEQ ID NO: 133, a CDR L2 as set forth in SEQ ID NO: 134, and a CDR L3 as set forth in SEQ ID NO: 135.
- the heavy chain sequence has the sequence of SEQ ID NO:247 and said light chain sequence has the sequence of SEQ ID NO:248.
- the antibody includes any one of the heavy chain sequences set forth by Table 9, 10, 11 or 12. In embodiments, the antibody includes any one of the heavy chain sequences set forth by Table 9. In embodiments, the antibody includes any one of the heavy chain sequences set forth by Table 10. In embodiments, the antibody includes any one of the heavy chain sequences set forth by Table 11. In embodiments, the antibody includes any one of the heavy chain sequences set forth by Table 12. In embodiments, the antibody includes any one of the light chain sequences set forth by Table 9, 10, 11 or 12. In embodiments, the antibody includes any one of the light chain sequences set forth by Table 9.
- the antibody includes any one of the light chain sequences set forth by Table 10. In embodiments, the antibody includes any one of the light chain sequences set forth by Table 11. In embodiments, the antibody includes any one of the light chain sequences set forth by Table 12. In embodiments, the antibody includes one of the heavy chain sequence and light chain sequence combinations set forth by Table 9, 10, 11 or 12. In embodiments, the antibody includes one of the heavy chain sequence and light chain sequence combinations set forth by Table 9. In embodiments, the antibody includes one of the heavy chain sequence and light chain sequence combinations set forth by Table 10. In embodiments, the antibody includes one of the heavy chain sequence and light chain sequence combinations set forth by Table 11. In embodiments, the antibody includes one of the heavy chain sequence and light chain sequence combinations set forth by Table 12.
- the anti-EGFR antibody is capable of binding to EGFR. In embodiments, the anti-EGFR antibody is capable of binding domain IV of EGFR. In embodiments, the anti-EGFR antibody does not substantially bind to domain I, domain II or domain III of EGFR.
- an antibody provided herein including embodiments thereof bound to domain IV of EGFR.
- a gene encoding a modified endogenous cell surface molecule may be used as a cell selection or enrichment marker for a genetically modified population of immune cells (e.g., T cells).
- the gene encoding a modified endogenous cell surface molecule may be coupled to a gene encoding a tumor targeting chimeric antigen receptor (CAR). These genes may be inserted into a vector to transduce the population of T cells to be genetically modified.
- the cells that are successfully transduced and express the CAR and modified endogenous cell-surface molecule are enriched by any suitable purification method, such as immunomagnetic purification with anti-biotin microbeads or fluorochrome-conjugated anti-biotin for fluorescence activated cell sorting, using a commercial antibody that recognizes the modified endogenous cell-surface molecule expressed by the transduced cell.
- a suitable purification method such as immunomagnetic purification with anti-biotin microbeads or fluorochrome-conjugated anti-biotin for fluorescence activated cell sorting, using a commercial antibody that recognizes the modified endogenous cell-surface molecule expressed by the transduced cell.
- a gene encoding a truncated human epidermal growth factor receptor that lacks the membrane distal EGF -binding domain and the cytoplasmic signaling tail, but retains domain IV EGFR recognized by any of the antibodies provided herein including embodiments thereof.
- the EGFRt may be coupled with chimeric antigen receptors specific for a tumor associated antigen.
- the tumor associated antigen may be CD19, CD20, or CD22, or any other tumor associated antigen, but is preferably CD19 (CD19CAR).
- the tumor associated antigen is followed by a C-terminal 2A cleavable linker and the coding sequence for EGFRt.
- the biotinylated-antibody may be used in conjunction with commercially available anti-biotin microbeads for the purpose of immunomagnetic purification of the tumor associated antigen/CAR-expressing transductants.
- the tumor associated antigen is CD19
- the product is CD19CAR-expressing transductants.
- the biotinylated-antibody may be used in conjunction with Fluorochrome-conjugated anti -biotin for fluorescence activated cell sorting.
- a recombinant nucleic acid including a sequence encoding a truncated EGFR (tEGFR) cell surface molecule, wherein the tEGFR cell surface molecule includes an EGFR domain IV and does not include an EGFR domain III.
- the tEGFR cell surface molecule does not include an EGFR domain I, an EGFR domain II, an EGFR juxtamembrane domain or an EGFR tyrosine kinase domain.
- the tEGFR cell surface molecule is non-immunogenic.
- the tEGFR cell surface molecule is a human tEGFR cell surface molecule.
- the tEGFR cell surface molecule has a sequence identity of at least 85% to the amino acid sequence of SEQ ID NO:276. In embodiments, the tEGFR cell surface molecule has a sequence identity of at least 86% to the amino acid sequence of SEQ ID NO:276. In embodiments, the tEGFR cell surface molecule has a sequence identity of at least 87% to the amino acid sequence of SEQ ID NO:276. In embodiments, the tEGFR cell surface molecule has a sequence identity of at least 88% to the amino acid sequence of SEQ ID NO:276. In embodiments, the tEGFR cell surface molecule has a sequence identity of at least 89% to the amino acid sequence of SEQ ID NO:276.
- the tEGFR cell surface molecule has a sequence identity of at least 90% to the amino acid sequence of SEQ ID NO:276. In embodiments, the tEGFR cell surface molecule has a sequence identity of at least 91% to the amino acid sequence of SEQ ID NO:276. In embodiments, the tEGFR cell surface molecule has a sequence identity of at least 92% to the amino acid sequence of SEQ ID NO:276. In embodiments, the tEGFR cell surface molecule has a sequence identity of at least 93% to the amino acid sequence of SEQ ID NO:276. In embodiments, the tEGFR cell surface molecule has a sequence identity of at least 94% to the amino acid sequence of SEQ ID NO:.
- the tEGFR cell surface molecule has a sequence identity of at least 95% to the amino acid sequence of SEQ ID NO:276. In embodiments, the tEGFR cell surface molecule has a sequence identity of at least 96% to the amino acid sequence of SEQ ID NO:276. In embodiments, the tEGFR cell surface molecule has a sequence identity of at least 97% to the amino acid sequence of SEQ ID NO:276. In embodiments, the tEGFR cell surface molecule has a sequence identity of at least 98% to the amino acid sequence of SEQ ID NO:276. In embodiments, the tEGFR cell surface molecule has a sequence identity of at least 99% to the amino acid sequence of SEQ ID NO:276.
- tEGFR cell surface molecule has a sequence identity of at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% to the amino acid sequence of SEQ ID NO:276.
- the tEGFR cell surface molecule binds an anti-domain IV EGFR antibody. In embodiments, the tEGFR cell surface molecule does not bind an anti-domain III EGFR antibody.
- the recombinant nucleic acid further includes a sequence encoding a chimeric antigen receptor, a T cell receptor, or a cytokine receptor. In embodiments, the recombinant nucleic acid further includes a sequence encoding a chimeric antigen receptor.
- the recombinant nucleic acid further includes a sequence encoding a T cell receptor. In embodiments, the recombinant nucleic acid further includes a sequence encoding a cytokine receptor.
- the chimeric antigen receptor includes an antibody region and a transmembrane domain.
- the antibody region binds to a cancer-antigen.
- the antibody region binds to CD 19.
- the sequence encoding the chimeric antigen receptor further includes an intracellular T-cell signaling domain.
- the cytokine receptor is IL15.
- the r ecombinant nucleic acid further includes a sequence encoding a self-cleaving peptidyl sequence.
- the self-cleaving peptidyl sequence connects the sequence encoding the tEGFR cell surface molecule with the sequence encoding the chimeric antigen receptor.
- the self-cleaving peptidyl sequence encodes a T2A peptidyl sequence, P2A peptidyl sequence, a E2A peptidyl sequence, a F2A peptidyl sequence or a 2A peptidyl sequence.
- the self cleaving peptidyl sequence encodes a T2A peptidyl sequence.
- the self cleaving peptidyl sequence encodes a P2A peptidyl sequence.
- the self cleaving peptidyl sequence encodes a E2A peptidyl sequence.
- the self cleaving peptidyl sequence encodes a F2A peptidyl sequence.
- the self cleaving peptidyl sequence encodes a 2A peptidyl sequence.
- an expression vector including the recombinant nucleic acid provided herein including embodiments thereof.
- the expression vector is an adenoviral vector or a retroviral vector.
- the expression vector is an adenoviral vector.
- the expression vector is a retroviral vector.
- a cell including a tEGFR cell surface molecule provided herein including embodiments thereof or an expression vector provided herein including embodiments thereof.
- the cell is bound to an anti-domain IV EGFR antibody in vitro or in vivo.
- the cell is bound to an anti-domain IV EGFR antibody in vitro.
- the cell is bound to an anti-domain IV EGFR antibody in vivo.
- the cell is a T cell, a natural killer (Nk) cell or an induced pluripotent stem cell (iPSC).
- the cell is a T cell.
- the cell is a natural killer (Nk) cell.
- the cell is an induced pluripotent stem cell (iPSC).
- the Nk cell is a cytoprotective Nk cell.
- the Nk cell is a cytotoxic NK cell.
- kit composition including (i) an expression vector provided herein including embodiments thereof; and (ii) an anti-domain IV EGFR antibody or an expression vector encoding an anti-domain IV EGFR antibody.
- the expression vector of (i) and the expression vector or said antibody of (ii) are in separate containers.
- a method of selecting a cell expressing a tEGFR cell surface molecule including: (i) contacting a population of cells with a recombinant nucleic acid provided herein including embodiments thereof or an expression vector provided herein including embodiments thereof, thereby forming a contacted cell population; (ii) contacting the contacted cell population with a tEGFR binding agent, thereby forming a bound tEGFR expressing cell; and (iii) separating the bound tEGFR expressing cell from the contacted cell population, thereby selecting a cell expressing a tEGFR cell surface molecule.
- the tEGFR binding agent is an anti-domain IV EGFR antibody.
- the antibody includes a detectable moiety.
- the population of cells is in a subject. In embodiments, the population of cells is in a tissue culture container.
- a method of detecting a cell expressing a tEGFR cell surface molecule including: (i) contacting a population of cells expressing a recombinant nucleic acid provided herein including embodiments thereof or an expression vector provided herein including embodiments thereof with a tEGFR binding agent, and (ii) detecting binding of the binding agent to a tEGFR cell surface molecule thereby detecting a cell expressing a tEGFR cell surface molecule.
- the tEGFR binding agent is an anti-domain IV EGFR antibody.
- the antibody includes a detectable moiety.
- Embodiment 1 A recombinant nucleic acid comprising a sequence encoding a truncated EGFR (tEGFR) cell surface molecule, wherein said tEGFR cell surface molecule comprises an EGFR domain IV and does not comprise an EGFR domain III.
- tEGFR truncated EGFR
- Embodiment 2 The recombinant nucleic acid of embodiment 1, wherein said tEGFR cell surface molecule does not comprise an EGFR domain I, an EGFR domain II, an EGFR juxtamembrane domain or an EGFR tyrosine kinase domain.
- Embodiment 3 The recombinant nucleic acid of embodiment 1 or 2, wherein said tEGFR cell surface molecule is non-immunogenic.
- Embodiment 4 The recombinant nucleic acid of any one of embodiments 1-3, wherein said tEGFR cell surface molecule is a human tEGFR cell surface molecule.
- Embodiment 5 The recombinant nucleic acid of any one of claims 1-4, wherein said tEGFR cell surface molecule has a sequence identity of at least 85% to the amino acid sequence of SEQ ID NO:276.
- Embodiment 6 The recombinant nucleic acid of any one of claims 1-5, wherein said tEGFR cell surface molecule binds an anti-domain IV EGFR antibody.
- Embodiment 7 The recombinant nucleic acid of any one of claims 1-6, wherein said tEGFR cell surface molecule does not bind an anti-domain III EGFR antibody.
- Embodiment 8 The recombinant nucleic acid of any one of claims 1-6, further comprising a sequence encoding a chimeric antigen receptor, a T cell receptor, or a cytokine receptor.
- Embodiment 9 The recombinant nucleic acid of claim 8, wherein said chimeric antigen receptor comprises an antibody region and a transmembrane domain.
- Embodiment 10 The recombinant nucleic acid of claim 9, wherein said antibody region binds to a cancer-antigen.
- Embodiment 11 The recombinant nucleic acid of claim 9, wherein said antibody region binds to CD 19.
- Embodiment 12 The recombinant nucleic acid of any one of claims 8-11, wherein said sequence encoding said chimeric antigen receptor further comprises an intracellular T- cell signaling domain.
- Embodiment 13 The recombinant nucleic acid of claim 8, wherein said cytokine receptor is IL15.
- Embodiment 14 The recombinant nucleic acid of any one of claims 8-12, further comprising a sequence encoding a self-cleaving peptidyl sequence.
- Embodiment 15 The recombinant nucleic acid of claim 14, wherein said self cleaving peptidyl sequence connects said sequence encoding said tEGFR cell surface molecule with said sequence encoding said chimeric antigen receptor.
- Embodiment 16 The recombinant nucleic acid of claim 14 or 15, wherein said self cleaving peptidyl sequence encodes a T2A peptidyl sequence, P2A peptidyl sequence, a E2A peptidyl sequence, a F2A peptidyl sequence or a 2A peptidyl sequence.
- Embodiment 17 An expression vector comprising the recombinant nucleic acid of one of claims 1-16.
- Embodiment 18 The expression vector of claim 17, wherein said expression vector is an adenoviral vector or a retroviral vector.
- Embodiment 19 The expression vector of claim 18, wherein said retroviral vector is a retroviral vector.
- Embodiment 20 A cell comprising a tEGFR cell surface molecule of any one of claims 1-16 or an expression vector of any one of claims 17-18.
- Embodiment 21 The cell of claim 20, wherein said cell is bound to an anti-domain IV EGFR antibody in vitro or in vivo.
- Embodiment 22 The cell of claim 20 or 21, wherein said cell is a T cell, a natural killer (Nk) cell or an induce pluripotent stem cell (iPSC).
- Embodiment 23 A kit composition comprising (i) an expression vector of any one of claims 17-18; and (ii) an anti-domain IV EGFR antibody or an expression vector encoding an anti-domain IV EGFR antibody.
- Embodiment 24 The kit of claim 23, wherein said expression vector of (i) and said expression vector or said antibody of (ii) are in separate containers.
- Embodiment 25 A method of selecting a cell expressing a tEGFR cell surface molecule, said method comprising: (i) contacting a population of cells with a recombinant nucleic acid of any one of claims 1-16 or an expression vector of any one of claims 17-19, thereby forming a contacted cell population; (ii) contacting said contacted cell population with a tEGFR binding agent, thereby forming a bound tEGFR expressing cell; and (iii) separating said bound tEGFR expressing cell from said contacted cell population, thereby selecting a cell expressing a tEGFR cell surface molecule.
- Embodiment 26 The method of claim 25, wherein said tEGFR binding agent is an anti domain IV EGFR antibody.
- Embodiment 27 The method of claim 26, wherein said antibody comprises a detectable moiety.
- Embodiment 28 The method of any one of claims 25-27, wherein said population of cells is in a subject.
- Embodiment 29 The method of any one of claims 25-27, wherein said population of cells is in a tissue culture container.
- Embodiment 30 A method of detecting a cell expressing a tEGFR cell surface molecule, said method comprising: (i) contacting a population of cells expressing a recombinant nucleic acid of any one of claims 1-15 or an expression vector of any one of claims 17-19 with a tEGFR binding agent, and (ii) detecting binding of said binding agent to a tEGFR cell surface molecule thereby detecting a cell expressing a tEGFR cell surface molecule.
- Embodiment 31 The method of claim 30, wherein said tEGFR binding agent is an anti domain IV EGFR antibody.
- Embodiment 32 The method of claim 31, wherein said antibody comprises a detectable moiety.
- Example 1 Development of EGFR Domain IV Targeting Antibodies
- Various EGFR-targeting biologies were investigated for their binding location on the EGFR target. Based on crystallographic data, the superposition of biologies to the extracellular domain indicate that nearly all biologies bind to domain III of the EGFR target (FIG. 2). This is consistent with the mode of action of the biologies, specifically, of EGF blockade as the ligand binds to domain III and is secured by a large conformational change leading to domain I covering the EGF ligand. Likewise, non-antibody EGFR-targeting therapeutics bind to domain III, operating similarly by inhibiting EGF binding (FIG. 3).
- trastuzumab which is a highly effective therapeutic targeting Her2 positive tumors, does not have an extracellular ligand.
- Trastuzumab binds domain IV of Her2, the juxtamembrane domain (FIG. 4).
- the mechanism of action of trastuzumab is distinct from cetuximab.
- results show that trastuzumab strongly potentiates Antibody-Dependent Cellular Cytotoxicity (ADCC). This is in contrast to the significantly weaker effect that cetuximab induces.
- ADCC Antibody-Dependent Cellular Cytotoxicity
- Applicants show that in embodiments the antibodies provided herein are specific to overexpressing EGFR cell lines which express low or no Her2 expression. This suggests that the antibodies provided herein will have high specificity and safety. That is, side effects that accompany other less specific antibody therapeutics can be avoided.
- Applicants show in animal studies that ADCC competent anti-domain IV EGFR mAh eradicates MDA-MB-468 tumor xenograft, a triple negative breast cancer (TNBC) cell line (e.g., no Her2 expression).
- TNBC triple negative breast cancer
- an ADCC-silent variant of the mAh e.g., swapping Fes that do not bind CD16 fails to halt tumor growth (e.g., similar to PBS).
- TNBC triple-negative breast cancer
- NSCLC non-small cell lung cancer
- Chimeric human EGFR domain IV proteins were constructed by fusing the extracellular EGFR domain IV, or domains II to IV with mouse IgG2a.Fc, designed as D4-IgG2a (SEQ ID NO:273) and EGFR-D4-IgG2a (SEQ ID NO:274), respectively (FIG. 6).
- the extracellular domain IV of human EGFR fused with human IgAl.Fc was designated as D4-IgA (SEQ ID NO: 275) (FIG. 6).
- the DNAs encoding these fusion proteins were synthesized and cloned into a mammalian expression vector by the manufacture (Twist Bioscience).
- the chimeric proteins were produced using an ExpiCHO expression system (Thermo Fisher Scientific). The procedures were followed according to the manufacturer’s manual.
- CHO cells were seeded at 3-4 x 10 6 cells/mL in fresh medium one day before transfection. On the next day, cells were adjusted to 6 x 10 6 cells/mL.
- 160 ug of DNA was added into 8 mL of OptiPROTM SFM and then mixed with 640 uL of ExpiFectamineTM diluted in 7.4 mL OptiPROTM SFM. The mixture was slowly added into the cell culture with gentle swirling, and cells were then cultured at 37 °C for 16-22 h.
- ExpiCHOTM Enhancer 1.2 mL
- ExpiCHOTM Feed 48 mL
- Cell supernatants were harvested by centrifugation at 4000 x g for 30 minutes and passed through 0.22-mm filters for protein purification.
- Protein A resins GE Healthcare
- CaptureSelectTM IgA Affinity Matrix Thermo Fisher Scientific
- Immunogen proteins were further purified with a Superdex 200 Increase 10/300 GL column (GE Healthcare) (FIG. 7).
- mice were immunized with complete Freund’s adjuvants (Sigma Aldrich) and subcutaneously injected into 10 Balb/c mice (The Jackson Laboratory), respectively. Fifty micrograms of proteins were injected for each mouse. After three weeks, mice received two subcutaneous injections of 50 ug fusion proteins emulsified with incomplete Freund’s adjuvants (Sigma Aldrich) in a two-week interval. Three days before spleen harvests,
- fusion proteins 10 ug were injected into mice via tail veins.
- Spleen cells were harvested and fused with mouse myeloma cell line FO (ATCC) at 1:1 ratio using PEG 1500 (Roche). The cell fusion procedures were followed according to the manufacturer’s manual. After fusion, cells were selected in complete DMEM medium containing hypoxanthine/aminopterin/thymidine (Thermo Fisher Scientific) and 10 % UltraCruz® Hybridoma Cloning Supplement (Santa Cruz) for 10-12 days. Hybridoma culture supernatants were screened for reacting to human EGFR-IgA proteins with ELISAs.
- VH/VL sequences were extracted using a Quick-RNA Microprep kit (Zymo Research).
- First-strand cDNAs were synthesized using a Superscript III First-Strand Synthesis System (Thermo Fisher Scientific).
- the VH and VL fragments were amplified by PCRs using a Mouse Ig-Primer Set (Millipore Sigma) and OneTaq 2X Master Mix (NEB).
- Amplified DNA fragments were purified using a DNA Clean-up kit (Zymo Research) and ligated into pGEM-T vectors (Promega) for sequencing.
- Example 3 Generation and Characterization of anti-Domain IV EGFR [0349]
- the chimeric anti-domain IV EGFR antibody clone 5C8 (EGFRD4-5C8), which includes murine variable domains (Fvs), was expressed and purified.
- EGFRD4- 5C8 was purified by size exclusion chromatography (FIG. 13 A), and the purified sample was characterized by both reducing and non-reducing gels. As expected, the non-reducing gel showed a single high molecular weight band, while the reducing gel showed two lower molecular weight bands at approximately 25 kDa and 50 kDa (FIG. 13B).
- EGFRD4- 5C8 Fab was tested for analyzed for binding to EGFR domain IV.
- EGFRD4-5C8 Fab, cetuximab Fab, and wild type traszumab samples were prepared in HBS-EP + running buffer.
- the samples were injected at concentrations of 300 nM, 100 nM, 30 nM, 10 nM, and 3 nM (FIG.s 15A-15C).
- the sensorgram curves were subsequently analyzed.
- Comparison of 5C8 and 5C8 Fab sensorgrams show that the 5C8 IgG antibody has a relatively longer off-rate. Significantly, the faster off-rate of the Fab may be beneficial for targeting EGFR over-expressing cancer cells, while avoiding healthy cells.
- Example 4 In Vitro and In Vivo Characterization of anti-Domain IV EGFR antibody 5C8 [0351] Further experiments to elucidate the mode of action of the anti-domain IV EGFR antibody clone 5C8 were conducted.
- the ovarian cancer cell line OVCAR3 was incubated with EGF (positive control), cetuximab, or the 5C8 antibody.
- Western blots were completed to detect the presence of phosphorylated EGFR, phosphorylated Akt, and b-actin (control) (FIG. 16).
- phosphorylation of EGFR was inhibited by cetuximab.
- 5C8 did not inhibit production of phosphorylated-EGFR confirming that 5C8 does not act by blocking EGF binding.
- phosphorylated Akt was very weakly detected upon incubation with 5C8 and none was detected with cetuximab treatment.
- ADCC antibody dependent cellular cytotoxicity
- Results show that the 5C8 antibody potently activates the NFAT pathway, as shown by the increase in luminescence. Since there was not significant ADCC observed for cetuximab, a second Jurkat T cell activation readout system was created. Applicants swapped CD 16 with a 158V variant, another CD 16 isoform found in humans and shown to bind to the Fc with higher affinity. Once again, T cell activation was tested using the anti-domain IV EGFR 5C8 mAh and cetuximab. While cetuximab induced T cell activation was improved using the 158V variant, ADCC effects were still more subtle with cetuximab than with the EGFRd4 5C8 antibody clone (FIG.s 19A-19E). Moreover, the results show that generally, cells with higher expression levels of EGFR display greater ADCC effects (FIG.s 19F and 19G).
- EGFR expression in various cancer lines was assessed by flow cytometry.
- MDA-MB-468, SKOV3, SW48, A549 and HCT116 cells were incubated with 10 ug/ml 5C8 or 10 ug/ml cetuximab for 30 min and subsequently washed.
- Cells were stained with anti-kappa- Alexa-647 secondary antibody, and the median fluorescence intensity of each cell line before and after antibody binding was assessed (FIG. 20).
- mice Female SCID mice (BALB/c-Igh* scid ) were subcutaneously injected with five million MDA-MB-468 breast cancer cells on day 1. The mice were then divided into 4 groups of 5 mice for intraperitoneal administration with either 5 mg/kg PBS, 5 mg/kg 5C8-IgGl, 5 mg/kg 5C8- IgG2a, or 5 mg/kg, cetuximab-IgG2a. The weight of each mouse was approximately 20 g, thus approximately 100 ug of antibody was administered per mouse. Standard model endpoints were assessed by tumor volumes (i.e.
- Additional antibody screening revealed further anti-domain IV EGFR antibody clones.
- the antibodies were purified by size exclusion chromatography and subsequently characterized by reducing and non-reducing gels (FIG.s 25A-25F).
- Several of the identified antibody clones (EGFRD4-7Ab, EGFRD4-28Ab, EGFRD4-30Ab, EGFRD4-31Ab, and EGFRD4-34Ab) could be expressed and purified, with the exception of EGFRD4-26.
- the antibody clones are further assessed for ADCC both in vitro and in vivo. Further, expression is optimized for the clones, and additional members that did not express are generated. Fab domains of the clones are generated to characterize monomeric binding affinity and for crystallography with EGFR domain IV.
- FITC-, PE- and PerCP-conjugated isotype controls PerCP-conjugated anti-CD8, FITC conjugated anti-CD4, PE-conjugated anti-IFN. gamma., PerCP-conjugated anti-CD45 and PE- conjugated streptavidin were obtained from BD Biosciences (San Jose, Calif.). Biotinylated anti- Fc was purchased from Jackson ImmunoResearch Laboratories, Inc. (Westgrove, Pa.). PE- conjugated anti-Biotin was purchased from Miltenyi Biotec (Auburn, Calif.). Biotinylated EGF was purchased from Molecular Probes. RTM. Invitrogen (Carlsbad, Calif.).
- PE-conjugated anti- EGFR was purchased from Abeam Inc. (Cambridge, Mass.). All antibodies and biotin-EGF were used according to the manufacturer's instructions. Flow cytometric data acquisition was performed on a FACScalibur (BD Biosciences), and the percentage of cells in a region of analysis was calculated using FCS Express V3 (De Novo Software, Los Angeles, Calif.).
- biotinylated-domain IV antibodies 200 mg of an anti-domain IV EGFR antibody is buffer exchanged (19 hours) to PBS (D-PBS, pH 7.5.+-.0.1) using a MidGee Hoop Cartridge (UFP-30-E-H42LA) with 527 mL.
- PBS D-PBS, pH 7.5.+-.0.1
- UFP-30-E-H42LA MidGee Hoop Cartridge
- the material at 2 mg/mL is then modified at a 20: 1 ratio using Sulfo-NHS-LC-Biotin in a reaction that is carried out for 1 hour at room temperature and then diafiltered to remove the excess biotin.
- biotinylated anti domain IV EGFR antibody is then buffer exchanged (18 hours) to PBS (D-PBS, pH 7.5.+-.0.1) using MidGee Hoop Cartridge (UFP-30-E-H42LA) with 533 mL. Glycerol is added to a final concentration of 20% and then the material is frozen in vials.
- PBS D-PBS, pH 7.5.+-.0.1
- UFP-30-E-H42LA MidGee Hoop Cartridge
- Glycerol is added to a final concentration of 20% and then the material is frozen in vials.
- CM culture media
- PBMC peripheral blood mononuclear cells
- CMV-specific cells are generated by stimulating T cells with 5 U/ml rhIL-2 (Chiron, Emeryville, Calif.) and autologous irradiated viral antigen presenting cells at a 4: 1 (responder: stimulator) ratio once a week for three weeks, using 10% human serum instead of FCS to avoid non-specific stimulation.
- the viral antigen presenting cells are derived from PBMC that had been genetically modified to express CMVpp65 antigen.
- PBMC are resuspended in nucleofection solution using the Human T cell Nucleofector kit (Amaxa Inc., Gaithersberg, Md.), and 5.times.l0.sup.7 cells are aliquoted into 0.2-cm cuvettes containing 10 .mu.g HygroR-pp 65_pEK (or pmaxGFP from Amaxa Inc., as a transfection control) in a final volume of 100 mu.L/cuvette, and electroporated using the Amaxa Nucleofector I (Amaxa Inc.), program U-14, after which cells are allowed to recover for 6 hours at 37. degree. C. prior to .gamma. -irradiation (1200 cGy).
- Amaxa Nucleofector I Amaxa Inc.
- the CD19CAR-T2A-EGFRt_epHIV7 (pJ02104) and CD 19C AR-T2 A-EGFRt-T2 A- IMPDH2dm_epHIV7 (pJ02111) lenti viral constructs contain a) the chimeric antigen receptor (CAR) sequences consisting of the V.sub.H and V.sub.L gene segments of the CD19-specific FmC63 mAh, an IgGl hinge-C.sub.H2-C.sub.H3, the transmembrane and cytoplasmic signaling domains of the costimulatory molecule CD28, and the cytoplasmic domain of the CD3.zeta.
- CAR chimeric antigen receptor
- Lentiviral transduction is carried out on T cells that are stimulated with either 30 ng/mL anti-CD3.
- epsilon. (OKT3; Ortho Biotech, Raritan, N.J.) (i.e., for Line A) or human CD3/CD28Dynal beads at a 1:10 ratio (i.e., for Lines B, C, D and E) and 25 U IL2/ml.
- Cells are cultured for up to 2 hours at 37. degree. C.
- RetroNectin.RTM. 50 ug/ml coated plates prior to addition of the lentivirus at an MOI of 3 and 5 .mu.g/ml polyybrene. After 4 hours, warm medium is added to triple to volume, and the cells are then washed and plated in fresh media after 48 hours.
- AutoMACS.TM. sorting of EGFRt-expressing cells is carried out with biotinylated anti-domain IV EGFR antibody and anti-biotin microbeads (Miltenyi Biotec) as per the manufacturer's instructions.
- Expansion of T cells in rapid expansion medium (REM) involves the incubation of 10. sup.6 T cells with 30 ng/mL anti-CD3. epsilon.
- LCL-OKT3 cells are generated by resuspending LCL in nucleofection solution using the Amaxa Nucleofector kit T, adding OKT3-2A-Hygromycin_pEK (pJ01609) plasmid at 5 .mu.g/10.sup.7 cells, and electroporating cells using the Amaxa Nucleofector I, program T-20.
- the resulting LCL-OKT3-2A-Hygro_pEK (cJ03987) are grown in CM containing 0.4 mg/ml hygromycin.
- the mouse myeloma line NS0 (gift from Andrew Raubitschek, City of Hope National Medical Center, Duarte, Calif.) is resuspended in nucleofection solution using the Nucleofector kit T (Amaxa Inc., Gaithersberg, Md.), CD19t- DHFRdm-2 A-IL 12_pEK (pJ01607) or GFP-IMPDH2dm-2A-IL15_pcDNA3.1(+) (pJ01043) plasmid is added at 5 . mu. g/5. times.10.sup.6 cells, and cells are electroporated using the Amaxa Nucleofector I, program T-27.
- NS0-CD19t-DHFRdm-2A-IL12_pEK cJ03935
- NS0-GFP NS0-GFP : IMPDH2-IL 15 (IL2ss)_pcDNA3.1(+) (cJ02096) are grown in DMEM (Irvine Scientific, Santa Ana, Calif.) supplemented with 10% heat-inactivated FCS, 25 mM HEPES, and 2 mM L-glutamine in the presence of either 0.05 uM methotrexate (MTX) or 6 mu.M mycophenolic acid (MPA).
- U251T-pp 65 are generated by lenti viral transduction of U251T with pp 65-2A-eGFP-ffluc_epHIV7 (pJ01928) at an MOI of 1.
- the resulting U251T-pp 65-2A-eGFP- ffluc_epHIV7 are then FACS sorted for the GFP.sup.+ population (cJ05058).
- the Daudi lymphoma line is purchased from ATCC and grown in media consisting of RPMI 1640 (Irvine Scientific), 2 mM L-Glutamine (Irvine Scientific), 10% heat-inactivated FCS (Hyclone).
- SupB15 acute lymphoblastic leukemia cells and A431 epidermoid carcinoma cells are purchased from ATCC.
- Cells (up to 10. sup.7) are lysed with 80 mu.L of 1% Triton-X lysis buffer containing phosphatase inhibitor cocktail II (Sigma- Aldrich Corp., St. Louis, Mo.) (1:20 of inhibitor to buffer by volume). 50 .mu.g of protein is loaded in each lane, and Western blots are probed with antibodies from the Phospho-EGF receptor antibody sampler kit (Cell Signaling Technology, Inc., Danvers, Mass.) followed by IRDye.TM. 680CW or 800CW conjugated goat anti-rabbit antibodies (LI-COR, Lincoln, Nebr.), as well as the IRDye.TM. 800 conjugated anti-beta-Actin antibody (LI-COR) as per the manufacturers' instructions. Blots are imaged on the Odyssey Infrared Imaging System (LI-COR).
- IRDye.TM. 680CW or 800CW conjugated goat anti-rabbit antibodies IRDye.TM. 680CW conjugated goat anti
- the cytolytic activity of T cells is determined by 4-hour chromium-release assay (CRA), where effector cells are seeded into triplicate wells of V-bottom 96-well micro-plates containing 5. times.10.sup.3 51Cr-labeled targeT cells (Na.sub.2.sup.51Cr0.sub.4; (5mCi/mL); Amersham Pharmacia, Piscataway, N.J.) at various E:T ratios in 200 uL of CM and incubated for 4 hours at 5% CO.sub.2, 37. degree. C.
- CRA 4-hour chromium-release assay
- Antibody dependent cell mediated cytotoxicity is determined by chromium release as above using 5.times.l0.sup.3 51Cr-labeled target cells that have been pre-incubated for 90 min with up to 10 .mu.g/mL of anti-domain IV EGFR antibody, washed and then co-incubated with 5. times.10.sup.5 freshly isolated PBMC.
- mice are injected i.v. on day 0 with 10.sup.7 T cells. 2. times.10. sup.7 irradiated (8000 rads) NS0- GFP:IMPDH2-IL15(IL2ss)_pcDNA3.1(+) (cJ02096) cells are administered i.p. 3 times a week starting on day 0 to provide a systemic supply of human IL-15 in vivo. Bone marrow is harvested from euthanized animals and analyzed by flow cytometry. Antibody dependent cell mediated cytotoxicity assays are performed to determine the activity of anti-domain IV EGFR antibody against EGFRt.sup.+ T cells.
- biotinylated- anti-domain IV EGFR antibody is generated to be used in conjunction with commercially available anti biotin microbeads and an AutoMACS.TM. separator (Miltenyi Biotec) (FIG. 2c).
- Lentiviral transduction of various T cell lines with EGFRt-containing constructs, where the EGFRt gene is separated from other genes of interest on either one or both ends with the self-cleaving T2A sequence are generated. Surface detection may be accomplished with a EGFRt-sr39TK fusion. Immunomagnetic selection will allow for recovery of EGFRt.sup.+ T cell populations with greater than 90% purity.
- SEQ ID NO: 274 (EGFR-D4-IgG2a)
- GHEALPLAFTQKTIDRLAGKPTHVNVSVVMAEVDGTSY [0396] SEQ ID NO:276 (EGFR domain IV)
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Immunology (AREA)
- Molecular Biology (AREA)
- Organic Chemistry (AREA)
- Cell Biology (AREA)
- General Health & Medical Sciences (AREA)
- Engineering & Computer Science (AREA)
- Medicinal Chemistry (AREA)
- Biochemistry (AREA)
- Genetics & Genomics (AREA)
- Urology & Nephrology (AREA)
- Biophysics (AREA)
- Hematology (AREA)
- Biomedical Technology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Food Science & Technology (AREA)
- Analytical Chemistry (AREA)
- Physics & Mathematics (AREA)
- General Physics & Mathematics (AREA)
- Pathology (AREA)
- Microbiology (AREA)
- Biotechnology (AREA)
- Oncology (AREA)
- Hospice & Palliative Care (AREA)
- Toxicology (AREA)
- Zoology (AREA)
- Gastroenterology & Hepatology (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
Abstract
L'invention concerne une molécule de surface cellulaire d'EGFR tronqué (tEGFR) et des utilisations correspondantes. La molécule de surface cellulaire tEGFR comprend un domaine IV d'EGFR et ne comprend pas de domaine III d'EGFR et peut être utilisée, entre autres, en tant que marqueur de suivi in vivo pour des lymphocytes T humains génétiquement modifiés. En outre, la molécule de surface cellulaire tEGFR présente un potentiel d'épuisement cellulaire médié par l'intermédiaire d'anticorps spécifiques anti-domaine IV d'EGFR. Ainsi, les molécules de surface cellulaire tEGFR utilisées ici peuvent, entre autres, être utilisées comme outil de sélection non immunogène, marqueur de suivi, outil d'épuisement ou gène suicide pour des cellules génétiquement modifiées présentant un potentiel thérapeutique.
Priority Applications (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
PCT/US2022/016672 WO2022178040A1 (fr) | 2021-02-16 | 2022-02-16 | Egfr tronqué à domaine iv et ses utilisations |
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US202163150064P | 2021-02-16 | 2021-02-16 | |
US63/150,064 | 2021-02-16 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2022177677A1 true WO2022177677A1 (fr) | 2022-08-25 |
Family
ID=82931867
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2022/012621 WO2022177677A1 (fr) | 2021-02-16 | 2022-01-14 | Egfr de domaine iv tronqué et utilisations correspondantes |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2022177677A1 (fr) |
Cited By (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2024051762A1 (fr) * | 2022-09-07 | 2024-03-14 | Xadcera Biopharmaceutical (Suzhou) Co., Ltd. | Anticorps anti-trop2/egfr et leurs utilisations |
WO2024196798A1 (fr) * | 2023-03-17 | 2024-09-26 | Asher Biotherapeutics, Inc. | Protéine de fusion et son utilisation |
Citations (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20100239587A1 (en) * | 2001-04-06 | 2010-09-23 | The Trustees Of The University Of Pennsylvania | ErbB Interface Peptidomimetics And Methods Of Use Thereof |
US20150184128A1 (en) * | 2009-11-03 | 2015-07-02 | City Of Hope | TRUNCATED EPIDERIMAL GROWTH FACTOR RECEPTOR (EGFRt) FOR TRANSDUCED T CELL SELECTION |
WO2018226897A1 (fr) * | 2017-06-07 | 2018-12-13 | Intrexon Corporation | Expression de nouvelles étiquettes de cellules |
-
2022
- 2022-01-14 WO PCT/US2022/012621 patent/WO2022177677A1/fr active Application Filing
Patent Citations (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20100239587A1 (en) * | 2001-04-06 | 2010-09-23 | The Trustees Of The University Of Pennsylvania | ErbB Interface Peptidomimetics And Methods Of Use Thereof |
US20150184128A1 (en) * | 2009-11-03 | 2015-07-02 | City Of Hope | TRUNCATED EPIDERIMAL GROWTH FACTOR RECEPTOR (EGFRt) FOR TRANSDUCED T CELL SELECTION |
WO2018226897A1 (fr) * | 2017-06-07 | 2018-12-13 | Intrexon Corporation | Expression de nouvelles étiquettes de cellules |
US20180362940A1 (en) * | 2017-06-07 | 2018-12-20 | Intrexon Corporation | Expression of novel cell tags |
Cited By (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2024051762A1 (fr) * | 2022-09-07 | 2024-03-14 | Xadcera Biopharmaceutical (Suzhou) Co., Ltd. | Anticorps anti-trop2/egfr et leurs utilisations |
WO2024196798A1 (fr) * | 2023-03-17 | 2024-09-26 | Asher Biotherapeutics, Inc. | Protéine de fusion et son utilisation |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
JP7069261B2 (ja) | Cd73特異的結合分子及びその使用 | |
JP7116126B2 (ja) | 形質導入T細胞選択のためのトランケート上皮増殖因子レセプタ(EGFRt) | |
EP3313874B1 (fr) | Lymphocytes t récepteurs antigéniques chimériques de masquage pour l'activation spécifique d'une tumeur | |
ES2818103T3 (es) | Anticuerpos y receptores de antígenos quiméricos específicos para ROR1 | |
EP2850106B1 (fr) | Immunofusion bispécifique (ifb) de scfv se liant au cd123 et cd3 | |
EP2884999B1 (fr) | Procédé et compositions pour l'immunothérapie cellulaire | |
JP7539161B2 (ja) | Hla-drに結合するキメラ抗原受容体およびcar-t細胞 | |
WO2022177677A1 (fr) | Egfr de domaine iv tronqué et utilisations correspondantes | |
JPWO2012176765A1 (ja) | 抗ヒトp−カドへリン(cdh3)遺伝子組み換え抗体 | |
CA2760642A1 (fr) | Anticorps anti-cadherine | |
JP2021533761A (ja) | Ox40結合性ポリペプチド及びその使用 | |
JP2023547380A (ja) | 新規の抗lilrb2抗体および誘導体生成物 | |
CN116547302A (zh) | 靶向egfr的抗体及其用途 | |
CN115698072A (zh) | 抗gpc3抗体,抗gpc3嵌合抗原受体和gpc3/cd3双特异性抗体 | |
CN110291107B (zh) | 靶向cd43的独特唾液酸糖基化的癌症相关表位的单克隆抗体 | |
WO2022178040A1 (fr) | Egfr tronqué à domaine iv et ses utilisations | |
TW202313699A (zh) | 新型抗sirpa抗體 | |
EP4200325A1 (fr) | Région fc modifiée non naturelle humaine d'igg se liant spécifiquement à un récepteur fc modifié non naturel | |
WO2022029496A1 (fr) | Anticorps bispécifiques anti-her2 / anti-4-1bb et leurs utilisations | |
US20240228629A1 (en) | Anti-domain iv egfr antibodies and uses thereof | |
CN114773485B (zh) | 抗人PD-L1抗体和TGFβRII的双功能融合蛋白分子 | |
US20240368304A1 (en) | Humanized f77 antibodies and fragments thereof | |
RU2822461C1 (ru) | Трансгенные генетические метки и способы применения | |
AU2023228374A1 (en) | Therapeutic antibodies | |
WO2023244547A9 (fr) | Anticorps humanisés dirigés contre l'intégrine avb8 humaine |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 22756673 Country of ref document: EP Kind code of ref document: A1 |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
122 | Ep: pct application non-entry in european phase |
Ref document number: 22756673 Country of ref document: EP Kind code of ref document: A1 |