WO2024114687A1 - Anti-tfr1 antibodies and uses thereof - Google Patents
Anti-tfr1 antibodies and uses thereof Download PDFInfo
- Publication number
- WO2024114687A1 WO2024114687A1 PCT/CN2023/135130 CN2023135130W WO2024114687A1 WO 2024114687 A1 WO2024114687 A1 WO 2024114687A1 CN 2023135130 W CN2023135130 W CN 2023135130W WO 2024114687 A1 WO2024114687 A1 WO 2024114687A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- antibody
- antigen
- binding fragment
- vhh
- amino acid
- Prior art date
Links
- 230000027455 binding Effects 0.000 claims abstract description 249
- 239000000427 antigen Substances 0.000 claims abstract description 229
- 102000036639 antigens Human genes 0.000 claims abstract description 228
- 108091007433 antigens Proteins 0.000 claims abstract description 228
- 239000012634 fragment Substances 0.000 claims abstract description 169
- 108010033576 Transferrin Receptors Proteins 0.000 claims abstract description 9
- 102000007238 Transferrin Receptors Human genes 0.000 claims abstract 2
- 241000282414 Homo sapiens Species 0.000 claims description 103
- 108010047041 Complementarity Determining Regions Proteins 0.000 claims description 90
- 238000000034 method Methods 0.000 claims description 76
- 125000003275 alpha amino acid group Chemical group 0.000 claims description 73
- 102100026144 Transferrin receptor protein 1 Human genes 0.000 claims description 65
- 210000004027 cell Anatomy 0.000 claims description 60
- 229940049595 antibody-drug conjugate Drugs 0.000 claims description 42
- 210000004556 brain Anatomy 0.000 claims description 37
- 239000003814 drug Substances 0.000 claims description 31
- 229940124597 therapeutic agent Drugs 0.000 claims description 27
- 239000013598 vector Substances 0.000 claims description 25
- 239000000203 mixture Substances 0.000 claims description 24
- 150000007523 nucleic acids Chemical class 0.000 claims description 23
- 108091033319 polynucleotide Proteins 0.000 claims description 21
- 239000002157 polynucleotide Substances 0.000 claims description 21
- 102000040430 polynucleotide Human genes 0.000 claims description 21
- 239000000611 antibody drug conjugate Substances 0.000 claims description 18
- 239000008194 pharmaceutical composition Substances 0.000 claims description 18
- 208000014644 Brain disease Diseases 0.000 claims description 17
- 239000003795 chemical substances by application Substances 0.000 claims description 15
- 102000039446 nucleic acids Human genes 0.000 claims description 15
- 108020004707 nucleic acids Proteins 0.000 claims description 15
- 206010028980 Neoplasm Diseases 0.000 claims description 14
- 230000008499 blood brain barrier function Effects 0.000 claims description 14
- 210000001218 blood-brain barrier Anatomy 0.000 claims description 14
- 208000003174 Brain Neoplasms Diseases 0.000 claims description 13
- 201000011510 cancer Diseases 0.000 claims description 10
- 241000282693 Cercopithecidae Species 0.000 claims description 8
- 125000000613 asparagine group Chemical group N[C@@H](CC(N)=O)C(=O)* 0.000 claims description 6
- 239000003937 drug carrier Substances 0.000 claims description 5
- 235000004279 alanine Nutrition 0.000 claims description 4
- 210000004978 chinese hamster ovary cell Anatomy 0.000 claims description 3
- 238000012258 culturing Methods 0.000 claims description 3
- 206010006187 Breast cancer Diseases 0.000 claims description 2
- 208000026310 Breast neoplasm Diseases 0.000 claims description 2
- 206010009944 Colon cancer Diseases 0.000 claims description 2
- 208000001333 Colorectal Neoplasms Diseases 0.000 claims description 2
- 206010058467 Lung neoplasm malignant Diseases 0.000 claims description 2
- 206010033128 Ovarian cancer Diseases 0.000 claims description 2
- 206010061535 Ovarian neoplasm Diseases 0.000 claims description 2
- 206010060862 Prostate cancer Diseases 0.000 claims description 2
- 208000000236 Prostatic Neoplasms Diseases 0.000 claims description 2
- 208000005718 Stomach Neoplasms Diseases 0.000 claims description 2
- 125000003295 alanine group Chemical group N[C@@H](C)C(=O)* 0.000 claims description 2
- 239000002299 complementary DNA Substances 0.000 claims description 2
- 239000000824 cytostatic agent Substances 0.000 claims description 2
- 231100000433 cytotoxic Toxicity 0.000 claims description 2
- 239000002254 cytotoxic agent Substances 0.000 claims description 2
- 230000001472 cytotoxic effect Effects 0.000 claims description 2
- 206010017758 gastric cancer Diseases 0.000 claims description 2
- 208000032839 leukemia Diseases 0.000 claims description 2
- 201000007270 liver cancer Diseases 0.000 claims description 2
- 208000014018 liver neoplasm Diseases 0.000 claims description 2
- 201000005202 lung cancer Diseases 0.000 claims description 2
- 208000020816 lung neoplasm Diseases 0.000 claims description 2
- 201000011549 stomach cancer Diseases 0.000 claims description 2
- 235000001014 amino acid Nutrition 0.000 description 46
- 150000001413 amino acids Chemical group 0.000 description 45
- 229940024606 amino acid Drugs 0.000 description 44
- 238000006467 substitution reaction Methods 0.000 description 38
- 238000012217 deletion Methods 0.000 description 31
- 230000037430 deletion Effects 0.000 description 31
- 238000003780 insertion Methods 0.000 description 30
- 230000037431 insertion Effects 0.000 description 30
- 241000699670 Mus sp. Species 0.000 description 29
- 108090000765 processed proteins & peptides Proteins 0.000 description 27
- 108090000623 proteins and genes Proteins 0.000 description 27
- 102000004169 proteins and genes Human genes 0.000 description 25
- 102000004196 processed proteins & peptides Human genes 0.000 description 24
- 229920001184 polypeptide Polymers 0.000 description 22
- 235000018102 proteins Nutrition 0.000 description 22
- 239000003112 inhibitor Substances 0.000 description 19
- XEEYBQQBJWHFJM-UHFFFAOYSA-N Iron Chemical compound [Fe] XEEYBQQBJWHFJM-UHFFFAOYSA-N 0.000 description 18
- 108010021625 Immunoglobulin Fragments Proteins 0.000 description 17
- 102000008394 Immunoglobulin Fragments Human genes 0.000 description 17
- 108060003951 Immunoglobulin Proteins 0.000 description 16
- 102000018358 immunoglobulin Human genes 0.000 description 16
- 238000011282 treatment Methods 0.000 description 16
- 102100033400 4F2 cell-surface antigen heavy chain Human genes 0.000 description 14
- 101000800023 Homo sapiens 4F2 cell-surface antigen heavy chain Proteins 0.000 description 14
- 230000001965 increasing effect Effects 0.000 description 14
- 238000001990 intravenous administration Methods 0.000 description 14
- 108020004414 DNA Proteins 0.000 description 11
- 241000699666 Mus <mouse, genus> Species 0.000 description 11
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 11
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 11
- 241001465754 Metazoa Species 0.000 description 10
- 108091028043 Nucleic acid sequence Proteins 0.000 description 10
- 201000010099 disease Diseases 0.000 description 10
- 238000002474 experimental method Methods 0.000 description 10
- 230000006870 function Effects 0.000 description 10
- NFGXHKASABOEEW-UHFFFAOYSA-N 1-methylethyl 11-methoxy-3,7,11-trimethyl-2,4-dodecadienoate Chemical compound COC(C)(C)CCCC(C)CC=CC(C)=CC(=O)OC(C)C NFGXHKASABOEEW-UHFFFAOYSA-N 0.000 description 9
- 239000000872 buffer Substances 0.000 description 9
- 238000000533 capillary isoelectric focusing Methods 0.000 description 9
- 230000003053 immunization Effects 0.000 description 9
- 230000003993 interaction Effects 0.000 description 9
- 229910052742 iron Inorganic materials 0.000 description 9
- 230000035772 mutation Effects 0.000 description 9
- 239000002773 nucleotide Substances 0.000 description 9
- 125000003729 nucleotide group Chemical group 0.000 description 9
- 230000001225 therapeutic effect Effects 0.000 description 9
- 239000012491 analyte Substances 0.000 description 8
- 210000004369 blood Anatomy 0.000 description 8
- 239000008280 blood Substances 0.000 description 8
- 230000000694 effects Effects 0.000 description 8
- 238000002649 immunization Methods 0.000 description 8
- 239000000523 sample Substances 0.000 description 8
- 230000008685 targeting Effects 0.000 description 8
- 238000003556 assay Methods 0.000 description 7
- 230000008901 benefit Effects 0.000 description 7
- 239000003623 enhancer Substances 0.000 description 7
- 239000013604 expression vector Substances 0.000 description 7
- 238000001727 in vivo Methods 0.000 description 7
- 238000002347 injection Methods 0.000 description 7
- 239000007924 injection Substances 0.000 description 7
- 230000004048 modification Effects 0.000 description 7
- 238000012986 modification Methods 0.000 description 7
- 239000011780 sodium chloride Substances 0.000 description 7
- 238000012360 testing method Methods 0.000 description 7
- WEVYAHXRMPXWCK-UHFFFAOYSA-N Acetonitrile Chemical compound CC#N WEVYAHXRMPXWCK-UHFFFAOYSA-N 0.000 description 6
- 208000024827 Alzheimer disease Diseases 0.000 description 6
- 206010012289 Dementia Diseases 0.000 description 6
- 241000700605 Viruses Species 0.000 description 6
- 230000002776 aggregation Effects 0.000 description 6
- 238000004220 aggregation Methods 0.000 description 6
- 125000000539 amino acid group Chemical group 0.000 description 6
- 238000004458 analytical method Methods 0.000 description 6
- -1 cationic lipid Chemical class 0.000 description 6
- 238000010494 dissociation reaction Methods 0.000 description 6
- 230000005593 dissociations Effects 0.000 description 6
- 238000011002 quantification Methods 0.000 description 6
- 210000002966 serum Anatomy 0.000 description 6
- 208000024891 symptom Diseases 0.000 description 6
- 241000124008 Mammalia Species 0.000 description 5
- 239000005557 antagonist Substances 0.000 description 5
- 238000004113 cell culture Methods 0.000 description 5
- 238000001514 detection method Methods 0.000 description 5
- 230000012202 endocytosis Effects 0.000 description 5
- 238000004519 manufacturing process Methods 0.000 description 5
- 102000005962 receptors Human genes 0.000 description 5
- 108020003175 receptors Proteins 0.000 description 5
- 239000000243 solution Substances 0.000 description 5
- 210000001519 tissue Anatomy 0.000 description 5
- 238000001890 transfection Methods 0.000 description 5
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 4
- 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 4
- 235000002198 Annona diversifolia Nutrition 0.000 description 4
- CIWBSHSKHKDKBQ-JLAZNSOCSA-N Ascorbic acid Chemical compound OC[C@H](O)[C@H]1OC(=O)C(O)=C1O CIWBSHSKHKDKBQ-JLAZNSOCSA-N 0.000 description 4
- 241000251730 Chondrichthyes Species 0.000 description 4
- 241000701022 Cytomegalovirus Species 0.000 description 4
- SHZGCJCMOBCMKK-UHFFFAOYSA-N D-mannomethylose Natural products CC1OC(O)C(O)C(O)C1O SHZGCJCMOBCMKK-UHFFFAOYSA-N 0.000 description 4
- 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 4
- 241000588724 Escherichia coli Species 0.000 description 4
- VTLYFUHAOXGGBS-UHFFFAOYSA-N Fe3+ Chemical compound [Fe+3] VTLYFUHAOXGGBS-UHFFFAOYSA-N 0.000 description 4
- PNNNRSAQSRJVSB-SLPGGIOYSA-N Fucose Natural products C[C@H](O)[C@@H](O)[C@H](O)[C@H](O)C=O PNNNRSAQSRJVSB-SLPGGIOYSA-N 0.000 description 4
- 241000282412 Homo Species 0.000 description 4
- SHZGCJCMOBCMKK-DHVFOXMCSA-N L-fucopyranose Chemical compound C[C@@H]1OC(O)[C@@H](O)[C@H](O)[C@@H]1O SHZGCJCMOBCMKK-DHVFOXMCSA-N 0.000 description 4
- 241000283984 Rodentia Species 0.000 description 4
- 102000004338 Transferrin Human genes 0.000 description 4
- 108090000901 Transferrin Proteins 0.000 description 4
- 239000000556 agonist Substances 0.000 description 4
- 230000009830 antibody antigen interaction Effects 0.000 description 4
- 238000012575 bio-layer interferometry Methods 0.000 description 4
- 230000000903 blocking effect Effects 0.000 description 4
- 230000007423 decrease Effects 0.000 description 4
- 238000011161 development Methods 0.000 description 4
- 239000012636 effector Substances 0.000 description 4
- 230000036541 health Effects 0.000 description 4
- 210000005154 hemibrain Anatomy 0.000 description 4
- 230000000670 limiting effect Effects 0.000 description 4
- 230000014759 maintenance of location Effects 0.000 description 4
- 230000001404 mediated effect Effects 0.000 description 4
- 239000013642 negative control Substances 0.000 description 4
- 239000008363 phosphate buffer Substances 0.000 description 4
- 239000002953 phosphate buffered saline Substances 0.000 description 4
- 238000011160 research Methods 0.000 description 4
- 230000028327 secretion Effects 0.000 description 4
- 238000002198 surface plasmon resonance spectroscopy Methods 0.000 description 4
- 239000012581 transferrin Substances 0.000 description 4
- 210000004881 tumor cell Anatomy 0.000 description 4
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Chemical compound O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 4
- WVDDGKGOMKODPV-UHFFFAOYSA-N Benzyl alcohol Chemical compound OCC1=CC=CC=C1 WVDDGKGOMKODPV-UHFFFAOYSA-N 0.000 description 3
- 108010019670 Chimeric Antigen Receptors Proteins 0.000 description 3
- 241000699800 Cricetinae Species 0.000 description 3
- 229920002307 Dextran Polymers 0.000 description 3
- 102000004190 Enzymes Human genes 0.000 description 3
- 108090000790 Enzymes Proteins 0.000 description 3
- 102000006496 Immunoglobulin Heavy Chains Human genes 0.000 description 3
- 108010019476 Immunoglobulin Heavy Chains Proteins 0.000 description 3
- 108010067060 Immunoglobulin Variable Region Proteins 0.000 description 3
- 102000017727 Immunoglobulin Variable Region Human genes 0.000 description 3
- 239000003798 L01XE11 - Pazopanib Substances 0.000 description 3
- 241000282838 Lama Species 0.000 description 3
- DNIAPMSPPWPWGF-UHFFFAOYSA-N Propylene glycol Chemical compound CC(O)CO DNIAPMSPPWPWGF-UHFFFAOYSA-N 0.000 description 3
- 241000700159 Rattus Species 0.000 description 3
- 239000002671 adjuvant Substances 0.000 description 3
- 230000001580 bacterial effect Effects 0.000 description 3
- 150000001875 compounds Chemical class 0.000 description 3
- 230000001054 cortical effect Effects 0.000 description 3
- 230000003247 decreasing effect Effects 0.000 description 3
- 238000001943 fluorescence-activated cell sorting Methods 0.000 description 3
- 230000013595 glycosylation Effects 0.000 description 3
- 238000006206 glycosylation reaction Methods 0.000 description 3
- 230000002163 immunogen Effects 0.000 description 3
- 238000000338 in vitro Methods 0.000 description 3
- 239000007788 liquid Substances 0.000 description 3
- 239000000463 material Substances 0.000 description 3
- 238000005259 measurement Methods 0.000 description 3
- 210000004925 microvascular endothelial cell Anatomy 0.000 description 3
- 210000004088 microvessel Anatomy 0.000 description 3
- 238000011275 oncology therapy Methods 0.000 description 3
- 229960001592 paclitaxel Drugs 0.000 description 3
- CUIHSIWYWATEQL-UHFFFAOYSA-N pazopanib Chemical compound C1=CC2=C(C)N(C)N=C2C=C1N(C)C(N=1)=CC=NC=1NC1=CC=C(C)C(S(N)(=O)=O)=C1 CUIHSIWYWATEQL-UHFFFAOYSA-N 0.000 description 3
- 230000035515 penetration Effects 0.000 description 3
- 238000002360 preparation method Methods 0.000 description 3
- 238000000746 purification Methods 0.000 description 3
- 239000000126 substance Substances 0.000 description 3
- 235000000346 sugar Nutrition 0.000 description 3
- RCINICONZNJXQF-MZXODVADSA-N taxol Chemical compound O([C@@H]1[C@@]2(C[C@@H](C(C)=C(C2(C)C)[C@H](C([C@]2(C)[C@@H](O)C[C@H]3OC[C@]3([C@H]21)OC(C)=O)=O)OC(=O)C)OC(=O)[C@H](O)[C@@H](NC(=O)C=1C=CC=CC=1)C=1C=CC=CC=1)O)C(=O)C1=CC=CC=C1 RCINICONZNJXQF-MZXODVADSA-N 0.000 description 3
- 108010058566 130-nm albumin-bound paclitaxel Proteins 0.000 description 2
- HZAXFHJVJLSVMW-UHFFFAOYSA-N 2-Aminoethan-1-ol Chemical compound NCCO HZAXFHJVJLSVMW-UHFFFAOYSA-N 0.000 description 2
- 102100033793 ALK tyrosine kinase receptor Human genes 0.000 description 2
- 101710168331 ALK tyrosine kinase receptor Proteins 0.000 description 2
- 108010029445 Agammaglobulinaemia Tyrosine Kinase Proteins 0.000 description 2
- DCXYFEDJOCDNAF-UHFFFAOYSA-N Asparagine Natural products OC(=O)C(N)CC(N)=O DCXYFEDJOCDNAF-UHFFFAOYSA-N 0.000 description 2
- 241000894006 Bacteria Species 0.000 description 2
- 239000012591 Dulbecco’s Phosphate Buffered Saline Substances 0.000 description 2
- HKVAMNSJSFKALM-GKUWKFKPSA-N Everolimus Chemical compound C1C[C@@H](OCCO)[C@H](OC)C[C@@H]1C[C@@H](C)[C@H]1OC(=O)[C@@H]2CCCCN2C(=O)C(=O)[C@](O)(O2)[C@H](C)CC[C@H]2C[C@H](OC)/C(C)=C/C=C/C=C/[C@@H](C)C[C@@H](C)C(=O)[C@H](OC)[C@H](O)/C(C)=C/[C@@H](C)C(=O)C1 HKVAMNSJSFKALM-GKUWKFKPSA-N 0.000 description 2
- WSFSSNUMVMOOMR-UHFFFAOYSA-N Formaldehyde Chemical compound O=C WSFSSNUMVMOOMR-UHFFFAOYSA-N 0.000 description 2
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 2
- DHMQDGOQFOQNFH-UHFFFAOYSA-N Glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 description 2
- 239000007995 HEPES buffer Substances 0.000 description 2
- 241000238631 Hexapoda Species 0.000 description 2
- 102000013463 Immunoglobulin Light Chains Human genes 0.000 description 2
- 108010065825 Immunoglobulin Light Chains Proteins 0.000 description 2
- 102000012011 Isocitrate Dehydrogenase Human genes 0.000 description 2
- 108010075869 Isocitrate Dehydrogenase Proteins 0.000 description 2
- QNAYBMKLOCPYGJ-REOHCLBHSA-N L-alanine Chemical compound C[C@H](N)C(O)=O QNAYBMKLOCPYGJ-REOHCLBHSA-N 0.000 description 2
- 241000282567 Macaca fascicularis Species 0.000 description 2
- 241000282339 Mustela Species 0.000 description 2
- 108091061960 Naked DNA Proteins 0.000 description 2
- 241000283973 Oryctolagus cuniculus Species 0.000 description 2
- 108091007960 PI3Ks Proteins 0.000 description 2
- ISWSIDIOOBJBQZ-UHFFFAOYSA-N Phenol Chemical compound OC1=CC=CC=C1 ISWSIDIOOBJBQZ-UHFFFAOYSA-N 0.000 description 2
- 102000003993 Phosphatidylinositol 3-kinases Human genes 0.000 description 2
- 108090000430 Phosphatidylinositol 3-kinases Proteins 0.000 description 2
- 108020005091 Replication Origin Proteins 0.000 description 2
- 241000714474 Rous sarcoma virus Species 0.000 description 2
- 240000004808 Saccharomyces cerevisiae Species 0.000 description 2
- 241000282898 Sus scrofa Species 0.000 description 2
- 108060008682 Tumor Necrosis Factor Proteins 0.000 description 2
- 102100029823 Tyrosine-protein kinase BTK Human genes 0.000 description 2
- 238000010521 absorption reaction Methods 0.000 description 2
- 210000005006 adaptive immune system Anatomy 0.000 description 2
- 238000010171 animal model Methods 0.000 description 2
- 239000002246 antineoplastic agent Substances 0.000 description 2
- 235000010323 ascorbic acid Nutrition 0.000 description 2
- 229960005070 ascorbic acid Drugs 0.000 description 2
- 239000011668 ascorbic acid Substances 0.000 description 2
- 235000009582 asparagine Nutrition 0.000 description 2
- 229960001230 asparagine Drugs 0.000 description 2
- 210000003719 b-lymphocyte Anatomy 0.000 description 2
- 230000015572 biosynthetic process Effects 0.000 description 2
- 230000037396 body weight Effects 0.000 description 2
- 210000004899 c-terminal region Anatomy 0.000 description 2
- 238000005119 centrifugation Methods 0.000 description 2
- 239000003153 chemical reaction reagent Substances 0.000 description 2
- OSASVXMJTNOKOY-UHFFFAOYSA-N chlorobutanol Chemical compound CC(C)(O)C(Cl)(Cl)Cl OSASVXMJTNOKOY-UHFFFAOYSA-N 0.000 description 2
- 238000004587 chromatography analysis Methods 0.000 description 2
- 239000011248 coating agent Substances 0.000 description 2
- 238000000576 coating method Methods 0.000 description 2
- 230000000295 complement effect Effects 0.000 description 2
- 230000008878 coupling Effects 0.000 description 2
- 238000010168 coupling process Methods 0.000 description 2
- 238000005859 coupling reaction Methods 0.000 description 2
- 125000000151 cysteine group Chemical group N[C@@H](CS)C(=O)* 0.000 description 2
- 238000007405 data analysis Methods 0.000 description 2
- 230000001419 dependent effect Effects 0.000 description 2
- 238000003745 diagnosis Methods 0.000 description 2
- 229940079593 drug Drugs 0.000 description 2
- 230000009977 dual effect Effects 0.000 description 2
- 238000004520 electroporation Methods 0.000 description 2
- 230000002255 enzymatic effect Effects 0.000 description 2
- 229960005167 everolimus Drugs 0.000 description 2
- 230000029142 excretion Effects 0.000 description 2
- 230000004927 fusion Effects 0.000 description 2
- 210000004602 germ cell Anatomy 0.000 description 2
- 210000005260 human cell Anatomy 0.000 description 2
- 210000004408 hybridoma Anatomy 0.000 description 2
- 230000002209 hydrophobic effect Effects 0.000 description 2
- 210000002865 immune cell Anatomy 0.000 description 2
- 238000010166 immunofluorescence Methods 0.000 description 2
- 238000011065 in-situ storage Methods 0.000 description 2
- 208000015181 infectious disease Diseases 0.000 description 2
- 238000001361 intraarterial administration Methods 0.000 description 2
- 238000007918 intramuscular administration Methods 0.000 description 2
- 238000007912 intraperitoneal administration Methods 0.000 description 2
- GOTYRUGSSMKFNF-UHFFFAOYSA-N lenalidomide Chemical compound C1C=2C(N)=CC=CC=2C(=O)N1C1CCC(=O)NC1=O GOTYRUGSSMKFNF-UHFFFAOYSA-N 0.000 description 2
- 229960004942 lenalidomide Drugs 0.000 description 2
- 239000003446 ligand Substances 0.000 description 2
- 230000003211 malignant effect Effects 0.000 description 2
- 239000011159 matrix material Substances 0.000 description 2
- 238000002844 melting Methods 0.000 description 2
- 230000008018 melting Effects 0.000 description 2
- 125000002496 methyl group Chemical group [H]C([H])([H])* 0.000 description 2
- 238000010172 mouse model Methods 0.000 description 2
- 230000007170 pathology Effects 0.000 description 2
- 229960000639 pazopanib Drugs 0.000 description 2
- WBXPDJSOTKVWSJ-ZDUSSCGKSA-L pemetrexed(2-) Chemical compound C=1NC=2NC(N)=NC(=O)C=2C=1CCC1=CC=C(C(=O)N[C@@H](CCC([O-])=O)C([O-])=O)C=C1 WBXPDJSOTKVWSJ-ZDUSSCGKSA-L 0.000 description 2
- 229920000642 polymer Polymers 0.000 description 2
- 239000013641 positive control Substances 0.000 description 2
- 230000008569 process Effects 0.000 description 2
- 238000000751 protein extraction Methods 0.000 description 2
- 230000006798 recombination Effects 0.000 description 2
- 230000001105 regulatory effect Effects 0.000 description 2
- 230000001177 retroviral effect Effects 0.000 description 2
- 239000012146 running buffer Substances 0.000 description 2
- 238000012163 sequencing technique Methods 0.000 description 2
- 241000894007 species Species 0.000 description 2
- 230000009870 specific binding Effects 0.000 description 2
- 238000007920 subcutaneous administration Methods 0.000 description 2
- 239000004094 surface-active agent Substances 0.000 description 2
- 238000002560 therapeutic procedure Methods 0.000 description 2
- 230000001988 toxicity Effects 0.000 description 2
- 231100000419 toxicity Toxicity 0.000 description 2
- 238000010361 transduction Methods 0.000 description 2
- 230000026683 transduction Effects 0.000 description 2
- 230000009261 transgenic effect Effects 0.000 description 2
- 238000013519 translation Methods 0.000 description 2
- 102000003390 tumor necrosis factor Human genes 0.000 description 2
- 241000701161 unidentified adenovirus Species 0.000 description 2
- 230000003612 virological effect Effects 0.000 description 2
- 210000005253 yeast cell Anatomy 0.000 description 2
- DIGQNXIGRZPYDK-WKSCXVIASA-N (2R)-6-amino-2-[[2-[[(2S)-2-[[2-[[(2R)-2-[[(2S)-2-[[(2R,3S)-2-[[2-[[(2S)-2-[[2-[[(2S)-2-[[(2S)-2-[[(2R)-2-[[(2S,3S)-2-[[(2R)-2-[[(2S)-2-[[(2S)-2-[[(2S)-2-[[2-[[(2S)-2-[[(2R)-2-[[2-[[2-[[2-[(2-amino-1-hydroxyethylidene)amino]-3-carboxy-1-hydroxypropylidene]amino]-1-hydroxy-3-sulfanylpropylidene]amino]-1-hydroxyethylidene]amino]-1-hydroxy-3-sulfanylpropylidene]amino]-1,3-dihydroxypropylidene]amino]-1-hydroxyethylidene]amino]-1-hydroxypropylidene]amino]-1,3-dihydroxypropylidene]amino]-1,3-dihydroxypropylidene]amino]-1-hydroxy-3-sulfanylpropylidene]amino]-1,3-dihydroxybutylidene]amino]-1-hydroxy-3-sulfanylpropylidene]amino]-1-hydroxypropylidene]amino]-1,3-dihydroxypropylidene]amino]-1-hydroxyethylidene]amino]-1,5-dihydroxy-5-iminopentylidene]amino]-1-hydroxy-3-sulfanylpropylidene]amino]-1,3-dihydroxybutylidene]amino]-1-hydroxy-3-sulfanylpropylidene]amino]-1,3-dihydroxypropylidene]amino]-1-hydroxyethylidene]amino]-1-hydroxy-3-sulfanylpropylidene]amino]-1-hydroxyethylidene]amino]hexanoic acid Chemical compound C[C@@H]([C@@H](C(=N[C@@H](CS)C(=N[C@@H](C)C(=N[C@@H](CO)C(=NCC(=N[C@@H](CCC(=N)O)C(=NC(CS)C(=N[C@H]([C@H](C)O)C(=N[C@H](CS)C(=N[C@H](CO)C(=NCC(=N[C@H](CS)C(=NCC(=N[C@H](CCCCN)C(=O)O)O)O)O)O)O)O)O)O)O)O)O)O)O)N=C([C@H](CS)N=C([C@H](CO)N=C([C@H](CO)N=C([C@H](C)N=C(CN=C([C@H](CO)N=C([C@H](CS)N=C(CN=C(C(CS)N=C(C(CC(=O)O)N=C(CN)O)O)O)O)O)O)O)O)O)O)O)O DIGQNXIGRZPYDK-WKSCXVIASA-N 0.000 description 1
- YXTKHLHCVFUPPT-YYFJYKOTSA-N (2s)-2-[[4-[(2-amino-5-formyl-4-oxo-1,6,7,8-tetrahydropteridin-6-yl)methylamino]benzoyl]amino]pentanedioic acid;(1r,2r)-1,2-dimethanidylcyclohexane;5-fluoro-1h-pyrimidine-2,4-dione;oxalic acid;platinum(2+) Chemical compound [Pt+2].OC(=O)C(O)=O.[CH2-][C@@H]1CCCC[C@H]1[CH2-].FC1=CNC(=O)NC1=O.C1NC=2NC(N)=NC(=O)C=2N(C=O)C1CNC1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 YXTKHLHCVFUPPT-YYFJYKOTSA-N 0.000 description 1
- ARBXEMIAJIJEQI-WDCZJNDASA-N (3s,4r,5r)-3,4-dihydroxy-5-(hydroxymethyl)piperidin-2-one Chemical compound OC[C@H]1CNC(=O)[C@@H](O)[C@@H]1O ARBXEMIAJIJEQI-WDCZJNDASA-N 0.000 description 1
- FDKXTQMXEQVLRF-ZHACJKMWSA-N (E)-dacarbazine Chemical compound CN(C)\N=N\c1[nH]cnc1C(N)=O FDKXTQMXEQVLRF-ZHACJKMWSA-N 0.000 description 1
- KKVYYGGCHJGEFJ-UHFFFAOYSA-N 1-n-(4-chlorophenyl)-6-methyl-5-n-[3-(7h-purin-6-yl)pyridin-2-yl]isoquinoline-1,5-diamine Chemical compound N=1C=CC2=C(NC=3C(=CC=CN=3)C=3C=4N=CNC=4N=CN=3)C(C)=CC=C2C=1NC1=CC=C(Cl)C=C1 KKVYYGGCHJGEFJ-UHFFFAOYSA-N 0.000 description 1
- IIZPXYDJLKNOIY-JXPKJXOSSA-N 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphocholine Chemical compound CCCCCCCCCCCCCCCC(=O)OC[C@H](COP([O-])(=O)OCC[N+](C)(C)C)OC(=O)CCC\C=C/C\C=C/C\C=C/C\C=C/CCCCC IIZPXYDJLKNOIY-JXPKJXOSSA-N 0.000 description 1
- UEJJHQNACJXSKW-UHFFFAOYSA-N 2-(2,6-dioxopiperidin-3-yl)-1H-isoindole-1,3(2H)-dione Chemical compound O=C1C2=CC=CC=C2C(=O)N1C1CCC(=O)NC1=O UEJJHQNACJXSKW-UHFFFAOYSA-N 0.000 description 1
- WAVYAFBQOXCGSZ-UHFFFAOYSA-N 2-fluoropyrimidine Chemical compound FC1=NC=CC=N1 WAVYAFBQOXCGSZ-UHFFFAOYSA-N 0.000 description 1
- AXRCEOKUDYDWLF-UHFFFAOYSA-N 3-(1-methyl-3-indolyl)-4-[1-[1-(2-pyridinylmethyl)-4-piperidinyl]-3-indolyl]pyrrole-2,5-dione Chemical compound C12=CC=CC=C2N(C)C=C1C(C(NC1=O)=O)=C1C(C1=CC=CC=C11)=CN1C(CC1)CCN1CC1=CC=CC=N1 AXRCEOKUDYDWLF-UHFFFAOYSA-N 0.000 description 1
- XXJWYDDUDKYVKI-UHFFFAOYSA-N 4-[(4-fluoro-2-methyl-1H-indol-5-yl)oxy]-6-methoxy-7-[3-(1-pyrrolidinyl)propoxy]quinazoline Chemical compound COC1=CC2=C(OC=3C(=C4C=C(C)NC4=CC=3)F)N=CN=C2C=C1OCCCN1CCCC1 XXJWYDDUDKYVKI-UHFFFAOYSA-N 0.000 description 1
- FWMNVWWHGCHHJJ-SKKKGAJSSA-N 4-amino-1-[(2r)-6-amino-2-[[(2r)-2-[[(2r)-2-[[(2r)-2-amino-3-phenylpropanoyl]amino]-3-phenylpropanoyl]amino]-4-methylpentanoyl]amino]hexanoyl]piperidine-4-carboxylic acid Chemical compound C([C@H](C(=O)N[C@H](CC(C)C)C(=O)N[C@H](CCCCN)C(=O)N1CCC(N)(CC1)C(O)=O)NC(=O)[C@H](N)CC=1C=CC=CC=1)C1=CC=CC=C1 FWMNVWWHGCHHJJ-SKKKGAJSSA-N 0.000 description 1
- NMUSYJAQQFHJEW-KVTDHHQDSA-N 5-azacytidine Chemical compound O=C1N=C(N)N=CN1[C@H]1[C@H](O)[C@H](O)[C@@H](CO)O1 NMUSYJAQQFHJEW-KVTDHHQDSA-N 0.000 description 1
- WXNSCLIZKHLNSG-MCZRLCSDSA-N 6-(2,5-dioxopyrrol-1-yl)-N-[2-[[2-[[(2S)-1-[[2-[[2-[[(10S,23S)-10-ethyl-18-fluoro-10-hydroxy-19-methyl-5,9-dioxo-8-oxa-4,15-diazahexacyclo[14.7.1.02,14.04,13.06,11.020,24]tetracosa-1,6(11),12,14,16,18,20(24)-heptaen-23-yl]amino]-2-oxoethoxy]methylamino]-2-oxoethyl]amino]-1-oxo-3-phenylpropan-2-yl]amino]-2-oxoethyl]amino]-2-oxoethyl]hexanamide Chemical compound CC[C@@]1(O)C(=O)OCC2=C1C=C1N(CC3=C1N=C1C=C(F)C(C)=C4CC[C@H](NC(=O)COCNC(=O)CNC(=O)[C@H](CC5=CC=CC=C5)NC(=O)CNC(=O)CNC(=O)CCCCCN5C(=O)C=CC5=O)C3=C14)C2=O WXNSCLIZKHLNSG-MCZRLCSDSA-N 0.000 description 1
- 206010069754 Acquired gene mutation Diseases 0.000 description 1
- 108010025188 Alcohol oxidase Proteins 0.000 description 1
- 108010083359 Antigen Receptors Proteins 0.000 description 1
- 102000006306 Antigen Receptors Human genes 0.000 description 1
- 102100021569 Apoptosis regulator Bcl-2 Human genes 0.000 description 1
- 101100339431 Arabidopsis thaliana HMGB2 gene Proteins 0.000 description 1
- 239000004475 Arginine Substances 0.000 description 1
- MLDQJTXFUGDVEO-UHFFFAOYSA-N BAY-43-9006 Chemical compound C1=NC(C(=O)NC)=CC(OC=2C=CC(NC(=O)NC=3C=C(C(Cl)=CC=3)C(F)(F)F)=CC=2)=C1 MLDQJTXFUGDVEO-UHFFFAOYSA-N 0.000 description 1
- 108091012583 BCL2 Proteins 0.000 description 1
- 241000283690 Bos taurus Species 0.000 description 1
- 102100032367 C-C motif chemokine 5 Human genes 0.000 description 1
- 102100025248 C-X-C motif chemokine 10 Human genes 0.000 description 1
- 102100036170 C-X-C motif chemokine 9 Human genes 0.000 description 1
- 125000001433 C-terminal amino-acid group Chemical group 0.000 description 1
- 229940045513 CTLA4 antagonist Drugs 0.000 description 1
- 241000282832 Camelidae Species 0.000 description 1
- 241000282836 Camelus dromedarius Species 0.000 description 1
- 241000282465 Canis Species 0.000 description 1
- 241000282472 Canis lupus familiaris Species 0.000 description 1
- 241000700198 Cavia Species 0.000 description 1
- 108091062157 Cis-regulatory element Proteins 0.000 description 1
- 102000005853 Clathrin Human genes 0.000 description 1
- 108010019874 Clathrin Proteins 0.000 description 1
- 102000008186 Collagen Human genes 0.000 description 1
- 108010035532 Collagen Proteins 0.000 description 1
- 208000035473 Communicable disease Diseases 0.000 description 1
- 241000557626 Corvus corax Species 0.000 description 1
- 239000004971 Cross linker Substances 0.000 description 1
- CMSMOCZEIVJLDB-UHFFFAOYSA-N Cyclophosphamide Chemical compound ClCCN(CCCl)P1(=O)NCCCO1 CMSMOCZEIVJLDB-UHFFFAOYSA-N 0.000 description 1
- FBPFZTCFMRRESA-FSIIMWSLSA-N D-Glucitol Natural products OC[C@H](O)[C@H](O)[C@@H](O)[C@H](O)CO FBPFZTCFMRRESA-FSIIMWSLSA-N 0.000 description 1
- FBPFZTCFMRRESA-KVTDHHQDSA-N D-Mannitol Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-KVTDHHQDSA-N 0.000 description 1
- FBPFZTCFMRRESA-JGWLITMVSA-N D-glucitol Chemical compound OC[C@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-JGWLITMVSA-N 0.000 description 1
- WQZGKKKJIJFFOK-QTVWNMPRSA-N D-mannopyranose Chemical group OC[C@H]1OC(O)[C@@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-QTVWNMPRSA-N 0.000 description 1
- 230000006820 DNA synthesis Effects 0.000 description 1
- BWGNESOTFCXPMA-UHFFFAOYSA-N Dihydrogen disulfide Chemical compound SS BWGNESOTFCXPMA-UHFFFAOYSA-N 0.000 description 1
- 241000255581 Drosophila <fruit fly, genus> Species 0.000 description 1
- 108050002772 E3 ubiquitin-protein ligase Mdm2 Proteins 0.000 description 1
- 102000012199 E3 ubiquitin-protein ligase Mdm2 Human genes 0.000 description 1
- 238000002965 ELISA Methods 0.000 description 1
- XXPXYPLPSDPERN-UHFFFAOYSA-N Ecteinascidin 743 Natural products COc1cc2C(NCCc2cc1O)C(=O)OCC3N4C(O)C5Cc6cc(C)c(OC)c(O)c6C(C4C(S)c7c(OC(=O)C)c(C)c8OCOc8c37)N5C XXPXYPLPSDPERN-UHFFFAOYSA-N 0.000 description 1
- 241000196324 Embryophyta Species 0.000 description 1
- 102100038132 Endogenous retrovirus group K member 6 Pro protein Human genes 0.000 description 1
- 101710121417 Envelope glycoprotein Proteins 0.000 description 1
- 241000283073 Equus caballus Species 0.000 description 1
- 241000701959 Escherichia virus Lambda Species 0.000 description 1
- 102100038595 Estrogen receptor Human genes 0.000 description 1
- 241000206602 Eukaryota Species 0.000 description 1
- 241000282324 Felis Species 0.000 description 1
- 241000282326 Felis catus Species 0.000 description 1
- 102100020997 Fractalkine Human genes 0.000 description 1
- 101710113436 GTPase KRas Proteins 0.000 description 1
- 108010010803 Gelatin Proteins 0.000 description 1
- 108700028146 Genetic Enhancer Elements Proteins 0.000 description 1
- 241000699694 Gerbillinae Species 0.000 description 1
- WQZGKKKJIJFFOK-GASJEMHNSA-N Glucose Natural products OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 1
- 239000004471 Glycine Substances 0.000 description 1
- 108090000288 Glycoproteins Proteins 0.000 description 1
- 102000003886 Glycoproteins Human genes 0.000 description 1
- 241000282575 Gorilla Species 0.000 description 1
- 108700010013 HMGB1 Proteins 0.000 description 1
- 101150021904 HMGB1 gene Proteins 0.000 description 1
- 102100037907 High mobility group protein B1 Human genes 0.000 description 1
- 101000797762 Homo sapiens C-C motif chemokine 5 Proteins 0.000 description 1
- 101000858088 Homo sapiens C-X-C motif chemokine 10 Proteins 0.000 description 1
- 101000947172 Homo sapiens C-X-C motif chemokine 9 Proteins 0.000 description 1
- 101000854520 Homo sapiens Fractalkine Proteins 0.000 description 1
- 101000840540 Homo sapiens Iduronate 2-sulfatase Proteins 0.000 description 1
- 101000599852 Homo sapiens Intercellular adhesion molecule 1 Proteins 0.000 description 1
- 101001050886 Homo sapiens Lysine-specific histone demethylase 1A Proteins 0.000 description 1
- 101000777293 Homo sapiens Serine/threonine-protein kinase Chk1 Proteins 0.000 description 1
- 101000766306 Homo sapiens Serotransferrin Proteins 0.000 description 1
- 101900082162 Human immunodeficiency virus type 1 group M subtype B Surface protein gp120 Proteins 0.000 description 1
- 102000009786 Immunoglobulin Constant Regions Human genes 0.000 description 1
- 108010009817 Immunoglobulin Constant Regions Proteins 0.000 description 1
- 102100025390 Integrin beta-2 Human genes 0.000 description 1
- 102100037877 Intercellular adhesion molecule 1 Human genes 0.000 description 1
- 108010002352 Interleukin-1 Proteins 0.000 description 1
- 108010002350 Interleukin-2 Proteins 0.000 description 1
- 108090000978 Interleukin-4 Proteins 0.000 description 1
- 239000002147 L01XE04 - Sunitinib Substances 0.000 description 1
- 239000005511 L01XE05 - Sorafenib Substances 0.000 description 1
- 239000002138 L01XE21 - Regorafenib Substances 0.000 description 1
- 239000002176 L01XE26 - Cabozantinib Substances 0.000 description 1
- 241000283953 Lagomorpha Species 0.000 description 1
- 241000282842 Lama glama Species 0.000 description 1
- 108010064548 Lymphocyte Function-Associated Antigen-1 Proteins 0.000 description 1
- 102100024985 Lysine-specific histone demethylase 1A Human genes 0.000 description 1
- 229930195725 Mannitol Natural products 0.000 description 1
- 108010061593 Member 14 Tumor Necrosis Factor Receptors Proteins 0.000 description 1
- 108010052285 Membrane Proteins Proteins 0.000 description 1
- 102000018697 Membrane Proteins Human genes 0.000 description 1
- 102000003792 Metallothionein Human genes 0.000 description 1
- 108090000157 Metallothionein Proteins 0.000 description 1
- 241000699660 Mus musculus Species 0.000 description 1
- 101100381978 Mus musculus Braf gene Proteins 0.000 description 1
- 101000969137 Mus musculus Metallothionein-1 Proteins 0.000 description 1
- 101100369221 Mus musculus Tfrc gene Proteins 0.000 description 1
- 230000004988 N-glycosylation Effects 0.000 description 1
- 229930193140 Neomycin Natural products 0.000 description 1
- 230000004989 O-glycosylation Effects 0.000 description 1
- 239000012823 PI3K/mTOR inhibitor Substances 0.000 description 1
- 229910019142 PO4 Inorganic materials 0.000 description 1
- 229930012538 Paclitaxel Natural products 0.000 description 1
- 241000282577 Pan troglodytes Species 0.000 description 1
- 229920002732 Polyanhydride Polymers 0.000 description 1
- 239000002202 Polyethylene glycol Substances 0.000 description 1
- 229920000954 Polyglycolide Polymers 0.000 description 1
- 229920001710 Polyorthoester Polymers 0.000 description 1
- 241000288906 Primates Species 0.000 description 1
- 101710100969 Receptor tyrosine-protein kinase erbB-3 Proteins 0.000 description 1
- 102100029986 Receptor tyrosine-protein kinase erbB-3 Human genes 0.000 description 1
- 241001068263 Replication competent viruses Species 0.000 description 1
- 241001068295 Replication defective viruses Species 0.000 description 1
- 235000014680 Saccharomyces cerevisiae Nutrition 0.000 description 1
- 241000293869 Salmonella enterica subsp. enterica serovar Typhimurium Species 0.000 description 1
- 229940119182 Selectin agonist Drugs 0.000 description 1
- 102100031081 Serine/threonine-protein kinase Chk1 Human genes 0.000 description 1
- 102100023085 Serine/threonine-protein kinase mTOR Human genes 0.000 description 1
- DWAQJAXMDSEUJJ-UHFFFAOYSA-M Sodium bisulfite Chemical compound [Na+].OS([O-])=O DWAQJAXMDSEUJJ-UHFFFAOYSA-M 0.000 description 1
- 241000256248 Spodoptera Species 0.000 description 1
- 241000187747 Streptomyces Species 0.000 description 1
- 210000001744 T-lymphocyte Anatomy 0.000 description 1
- 108010065917 TOR Serine-Threonine Kinases Proteins 0.000 description 1
- CBPNZQVSJQDFBE-FUXHJELOSA-N Temsirolimus Chemical compound C1C[C@@H](OC(=O)C(C)(CO)CO)[C@H](OC)C[C@@H]1C[C@@H](C)[C@H]1OC(=O)[C@@H]2CCCCN2C(=O)C(=O)[C@](O)(O2)[C@H](C)CC[C@H]2C[C@H](OC)/C(C)=C/C=C/C=C/[C@@H](C)C[C@@H](C)C(=O)[C@H](OC)[C@H](O)/C(C)=C/[C@@H](C)C(=O)C1 CBPNZQVSJQDFBE-FUXHJELOSA-N 0.000 description 1
- 108020005038 Terminator Codon Proteins 0.000 description 1
- 239000004098 Tetracycline Substances 0.000 description 1
- AYFVYJQAPQTCCC-UHFFFAOYSA-N Threonine Natural products CC(O)C(N)C(O)=O AYFVYJQAPQTCCC-UHFFFAOYSA-N 0.000 description 1
- 239000004473 Threonine Substances 0.000 description 1
- 108010022394 Threonine synthase Proteins 0.000 description 1
- 102000006601 Thymidine Kinase Human genes 0.000 description 1
- 108020004440 Thymidine kinase Proteins 0.000 description 1
- 102100023935 Transmembrane glycoprotein NMB Human genes 0.000 description 1
- 102100028785 Tumor necrosis factor receptor superfamily member 14 Human genes 0.000 description 1
- 206010046865 Vaccinia virus infection Diseases 0.000 description 1
- 241001416177 Vicugna pacos Species 0.000 description 1
- JXLYSJRDGCGARV-WWYNWVTFSA-N Vinblastine Natural products O=C(O[C@H]1[C@](O)(C(=O)OC)[C@@H]2N(C)c3c(cc(c(OC)c3)[C@]3(C(=O)OC)c4[nH]c5c(c4CCN4C[C@](O)(CC)C[C@H](C3)C4)cccc5)[C@@]32[C@H]2[C@@]1(CC)C=CCN2CC3)C JXLYSJRDGCGARV-WWYNWVTFSA-N 0.000 description 1
- IXKSXJFAGXLQOQ-XISFHERQSA-N WHWLQLKPGQPMY Chemical compound C([C@@H](C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CC(C)C)C(=O)N1CCC[C@H]1C(=O)NCC(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CC(O)=O)C(=O)N1CCC[C@H]1C(=O)N[C@@H](CCSC)C(=O)N[C@@H](CC=1C=CC(O)=CC=1)C(O)=O)NC(=O)[C@@H](N)CC=1C2=CC=CC=C2NC=1)C1=CNC=N1 IXKSXJFAGXLQOQ-XISFHERQSA-N 0.000 description 1
- 238000002835 absorbance Methods 0.000 description 1
- 150000001242 acetic acid derivatives Chemical class 0.000 description 1
- 230000002378 acidificating effect Effects 0.000 description 1
- 230000002411 adverse Effects 0.000 description 1
- 229940110282 alimta Drugs 0.000 description 1
- BFNBIHQBYMNNAN-UHFFFAOYSA-N ammonium sulfate Chemical compound N.N.OS(O)(=O)=O BFNBIHQBYMNNAN-UHFFFAOYSA-N 0.000 description 1
- 229910052921 ammonium sulfate Inorganic materials 0.000 description 1
- 235000011130 ammonium sulphate Nutrition 0.000 description 1
- 229960000723 ampicillin Drugs 0.000 description 1
- AVKUERGKIZMTKX-NJBDSQKTSA-N ampicillin Chemical compound C1([C@@H](N)C(=O)N[C@H]2[C@H]3SC([C@@H](N3C2=O)C(O)=O)(C)C)=CC=CC=C1 AVKUERGKIZMTKX-NJBDSQKTSA-N 0.000 description 1
- 239000003708 ampul Substances 0.000 description 1
- VJZITPJGSQKZMX-XDPRQOKASA-N amrubicin Chemical compound O([C@H]1C[C@](CC2=C(O)C=3C(=O)C4=CC=CC=C4C(=O)C=3C(O)=C21)(N)C(=O)C)[C@H]1C[C@H](O)[C@H](O)CO1 VJZITPJGSQKZMX-XDPRQOKASA-N 0.000 description 1
- 210000004102 animal cell Anatomy 0.000 description 1
- 239000003242 anti bacterial agent Substances 0.000 description 1
- 230000000844 anti-bacterial effect Effects 0.000 description 1
- 230000000259 anti-tumor effect Effects 0.000 description 1
- 230000010056 antibody-dependent cellular cytotoxicity Effects 0.000 description 1
- 239000003429 antifungal agent Substances 0.000 description 1
- 229940121375 antifungal agent Drugs 0.000 description 1
- 102000025171 antigen binding proteins Human genes 0.000 description 1
- 108091000831 antigen binding proteins Proteins 0.000 description 1
- 230000000890 antigenic effect Effects 0.000 description 1
- 239000003963 antioxidant agent Substances 0.000 description 1
- 235000006708 antioxidants Nutrition 0.000 description 1
- 230000009118 appropriate response Effects 0.000 description 1
- ODKSFYDXXFIFQN-UHFFFAOYSA-N arginine Natural products OC(=O)C(N)CCCNC(N)=N ODKSFYDXXFIFQN-UHFFFAOYSA-N 0.000 description 1
- 229960003005 axitinib Drugs 0.000 description 1
- RITAVMQDGBJQJZ-FMIVXFBMSA-N axitinib Chemical compound CNC(=O)C1=CC=CC=C1SC1=CC=C(C(\C=C\C=2N=CC=CC=2)=NN2)C2=C1 RITAVMQDGBJQJZ-FMIVXFBMSA-N 0.000 description 1
- 229960002756 azacitidine Drugs 0.000 description 1
- VSRXQHXAPYXROS-UHFFFAOYSA-N azanide;cyclobutane-1,1-dicarboxylic acid;platinum(2+) Chemical compound [NH2-].[NH2-].[Pt+2].OC(=O)C1(C(O)=O)CCC1 VSRXQHXAPYXROS-UHFFFAOYSA-N 0.000 description 1
- 239000011324 bead Substances 0.000 description 1
- 230000009286 beneficial effect Effects 0.000 description 1
- 235000019445 benzyl alcohol Nutrition 0.000 description 1
- 230000000975 bioactive effect Effects 0.000 description 1
- 229920000249 biocompatible polymer Polymers 0.000 description 1
- 230000033228 biological regulation Effects 0.000 description 1
- 238000006664 bond formation reaction Methods 0.000 description 1
- 210000004958 brain cell Anatomy 0.000 description 1
- DQXBYHZEEUGOBF-UHFFFAOYSA-N but-3-enoic acid;ethene Chemical compound C=C.OC(=O)CC=C DQXBYHZEEUGOBF-UHFFFAOYSA-N 0.000 description 1
- ONIQOQHATWINJY-UHFFFAOYSA-N cabozantinib Chemical compound C=12C=C(OC)C(OC)=CC2=NC=CC=1OC(C=C1)=CC=C1NC(=O)C1(C(=O)NC=2C=CC(F)=CC=2)CC1 ONIQOQHATWINJY-UHFFFAOYSA-N 0.000 description 1
- 229960001292 cabozantinib Drugs 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
- 239000002775 capsule Substances 0.000 description 1
- 150000001720 carbohydrates Chemical group 0.000 description 1
- 229960004562 carboplatin Drugs 0.000 description 1
- 108010021331 carfilzomib Proteins 0.000 description 1
- BLMPQMFVWMYDKT-NZTKNTHTSA-N carfilzomib Chemical compound C([C@@H](C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](CC(C)C)C(=O)[C@]1(C)OC1)NC(=O)CN1CCOCC1)CC1=CC=CC=C1 BLMPQMFVWMYDKT-NZTKNTHTSA-N 0.000 description 1
- 229960002438 carfilzomib Drugs 0.000 description 1
- 125000002091 cationic group Chemical group 0.000 description 1
- 229960002412 cediranib Drugs 0.000 description 1
- 230000004663 cell proliferation Effects 0.000 description 1
- 230000001413 cellular effect Effects 0.000 description 1
- 230000033077 cellular process Effects 0.000 description 1
- VERWOWGGCGHDQE-UHFFFAOYSA-N ceritinib Chemical compound CC=1C=C(NC=2N=C(NC=3C(=CC=CC=3)S(=O)(=O)C(C)C)C(Cl)=CN=2)C(OC(C)C)=CC=1C1CCNCC1 VERWOWGGCGHDQE-UHFFFAOYSA-N 0.000 description 1
- 230000008859 change Effects 0.000 description 1
- 239000002738 chelating agent Substances 0.000 description 1
- 229960004926 chlorobutanol Drugs 0.000 description 1
- 230000004087 circulation Effects 0.000 description 1
- DQLATGHUWYMOKM-UHFFFAOYSA-L cisplatin Chemical compound N[Pt](N)(Cl)Cl DQLATGHUWYMOKM-UHFFFAOYSA-L 0.000 description 1
- 229960004316 cisplatin Drugs 0.000 description 1
- 150000001860 citric acid derivatives Chemical class 0.000 description 1
- 229930193282 clathrin Natural products 0.000 description 1
- 238000003776 cleavage reaction Methods 0.000 description 1
- 229920001436 collagen Polymers 0.000 description 1
- 239000000562 conjugate Substances 0.000 description 1
- 238000013270 controlled release Methods 0.000 description 1
- 229960004397 cyclophosphamide Drugs 0.000 description 1
- 229960003901 dacarbazine Drugs 0.000 description 1
- 230000006378 damage Effects 0.000 description 1
- 230000002950 deficient Effects 0.000 description 1
- 230000001934 delay Effects 0.000 description 1
- 238000001212 derivatisation Methods 0.000 description 1
- 239000008121 dextrose Substances 0.000 description 1
- 235000005911 diet Nutrition 0.000 description 1
- 230000037213 diet Effects 0.000 description 1
- 230000004069 differentiation Effects 0.000 description 1
- 102000004419 dihydrofolate reductase Human genes 0.000 description 1
- UGMCXQCYOVCMTB-UHFFFAOYSA-K dihydroxy(stearato)aluminium Chemical compound CCCCCCCCCCCCCCCCCC(=O)O[Al](O)O UGMCXQCYOVCMTB-UHFFFAOYSA-K 0.000 description 1
- 239000012470 diluted sample Substances 0.000 description 1
- 239000003085 diluting agent Substances 0.000 description 1
- 239000000539 dimer Substances 0.000 description 1
- 229940042399 direct acting antivirals protease inhibitors Drugs 0.000 description 1
- 208000035475 disorder Diseases 0.000 description 1
- 238000009826 distribution Methods 0.000 description 1
- 230000006334 disulfide bridging Effects 0.000 description 1
- 231100000673 dose–response relationship Toxicity 0.000 description 1
- 229940121647 egfr inhibitor Drugs 0.000 description 1
- 210000001671 embryonic stem cell Anatomy 0.000 description 1
- 210000002472 endoplasmic reticulum Anatomy 0.000 description 1
- 210000002889 endothelial cell Anatomy 0.000 description 1
- 238000005516 engineering process Methods 0.000 description 1
- 229950002189 enzastaurin Drugs 0.000 description 1
- 238000011067 equilibration Methods 0.000 description 1
- 108010038795 estrogen receptors Proteins 0.000 description 1
- 239000005038 ethylene vinyl acetate Substances 0.000 description 1
- 210000003527 eukaryotic cell Anatomy 0.000 description 1
- 238000000684 flow cytometry Methods 0.000 description 1
- JYEFSHLLTQIXIO-SMNQTINBSA-N folfiri regimen Chemical compound FC1=CNC(=O)NC1=O.C1NC=2NC(N)=NC(=O)C=2N(C=O)C1CNC1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1.C1=C2C(CC)=C3CN(C(C4=C([C@@](C(=O)OC4)(O)CC)C=4)=O)C=4C3=NC2=CC=C1OC(=O)N(CC1)CCC1N1CCCCC1 JYEFSHLLTQIXIO-SMNQTINBSA-N 0.000 description 1
- 238000009472 formulation Methods 0.000 description 1
- 230000037433 frameshift Effects 0.000 description 1
- 230000033581 fucosylation Effects 0.000 description 1
- 230000002538 fungal effect Effects 0.000 description 1
- 108020001507 fusion proteins Proteins 0.000 description 1
- 102000037865 fusion proteins Human genes 0.000 description 1
- 229920000159 gelatin Polymers 0.000 description 1
- 239000008273 gelatin Substances 0.000 description 1
- 235000019322 gelatine Nutrition 0.000 description 1
- 235000011852 gelatine desserts Nutrition 0.000 description 1
- SDUQYLNIPVEERB-QPPQHZFASA-N gemcitabine Chemical compound O=C1N=C(N)C=CN1[C@H]1C(F)(F)[C@H](O)[C@@H](CO)O1 SDUQYLNIPVEERB-QPPQHZFASA-N 0.000 description 1
- 229960005277 gemcitabine Drugs 0.000 description 1
- 230000002068 genetic effect Effects 0.000 description 1
- 150000004676 glycans Chemical class 0.000 description 1
- 235000011187 glycerol Nutrition 0.000 description 1
- 230000012010 growth Effects 0.000 description 1
- 239000003481 heat shock protein 90 inhibitor Substances 0.000 description 1
- 238000010438 heat treatment Methods 0.000 description 1
- 238000004128 high performance liquid chromatography Methods 0.000 description 1
- 230000005745 host immune response Effects 0.000 description 1
- 238000003384 imaging method Methods 0.000 description 1
- 102000028557 immunoglobulin binding proteins Human genes 0.000 description 1
- 108091009323 immunoglobulin binding proteins Proteins 0.000 description 1
- 229940072221 immunoglobulins Drugs 0.000 description 1
- 230000003116 impacting effect Effects 0.000 description 1
- 239000007943 implant Substances 0.000 description 1
- 230000008676 import Effects 0.000 description 1
- 230000006872 improvement Effects 0.000 description 1
- 238000011534 incubation Methods 0.000 description 1
- 230000006698 induction Effects 0.000 description 1
- 230000001939 inductive effect Effects 0.000 description 1
- 230000002401 inhibitory effect Effects 0.000 description 1
- 230000000977 initiatory effect Effects 0.000 description 1
- 239000007972 injectable composition Substances 0.000 description 1
- 230000010354 integration Effects 0.000 description 1
- 230000002452 interceptive effect Effects 0.000 description 1
- 230000003834 intracellular effect Effects 0.000 description 1
- 239000007928 intraperitoneal injection Substances 0.000 description 1
- 238000010253 intravenous injection Methods 0.000 description 1
- JEIPFZHSYJVQDO-UHFFFAOYSA-N iron(III) oxide Inorganic materials O=[Fe]O[Fe]=O JEIPFZHSYJVQDO-UHFFFAOYSA-N 0.000 description 1
- 239000007951 isotonicity adjuster Substances 0.000 description 1
- 230000000366 juvenile effect Effects 0.000 description 1
- 230000002147 killing effect Effects 0.000 description 1
- 101150109249 lacI gene Proteins 0.000 description 1
- 101150066555 lacZ gene Proteins 0.000 description 1
- 150000002605 large molecules Chemical class 0.000 description 1
- 239000000787 lecithin Substances 0.000 description 1
- 235000010445 lecithin Nutrition 0.000 description 1
- 229940067606 lecithin Drugs 0.000 description 1
- 210000005240 left ventricle Anatomy 0.000 description 1
- 231100000518 lethal Toxicity 0.000 description 1
- 230000001665 lethal effect Effects 0.000 description 1
- 229920002521 macromolecule Polymers 0.000 description 1
- 210000004962 mammalian cell Anatomy 0.000 description 1
- 239000000594 mannitol Substances 0.000 description 1
- 235000010355 mannitol Nutrition 0.000 description 1
- 239000003550 marker Substances 0.000 description 1
- 238000001840 matrix-assisted laser desorption--ionisation time-of-flight mass spectrometry Methods 0.000 description 1
- 201000001441 melanoma Diseases 0.000 description 1
- 229920000609 methyl cellulose Polymers 0.000 description 1
- 235000010270 methyl p-hydroxybenzoate Nutrition 0.000 description 1
- 239000001923 methylcellulose Substances 0.000 description 1
- 235000010981 methylcellulose Nutrition 0.000 description 1
- 244000309715 mini pig Species 0.000 description 1
- 238000007479 molecular analysis Methods 0.000 description 1
- 239000000178 monomer Substances 0.000 description 1
- 201000002273 mucopolysaccharidosis II Diseases 0.000 description 1
- 208000022018 mucopolysaccharidosis type 2 Diseases 0.000 description 1
- LBWFXVZLPYTWQI-IPOVEDGCSA-N n-[2-(diethylamino)ethyl]-5-[(z)-(5-fluoro-2-oxo-1h-indol-3-ylidene)methyl]-2,4-dimethyl-1h-pyrrole-3-carboxamide;(2s)-2-hydroxybutanedioic acid Chemical compound OC(=O)[C@@H](O)CC(O)=O.CCN(CC)CCNC(=O)C1=C(C)NC(\C=C/2C3=CC(F)=CC=C3NC\2=O)=C1C LBWFXVZLPYTWQI-IPOVEDGCSA-N 0.000 description 1
- 229960004927 neomycin Drugs 0.000 description 1
- 210000002569 neuron Anatomy 0.000 description 1
- 239000000346 nonvolatile oil Substances 0.000 description 1
- 238000011580 nude mouse model Methods 0.000 description 1
- 238000005457 optimization Methods 0.000 description 1
- 210000000056 organ Anatomy 0.000 description 1
- 238000004806 packaging method and process Methods 0.000 description 1
- 229960004390 palbociclib Drugs 0.000 description 1
- AHJRHEGDXFFMBM-UHFFFAOYSA-N palbociclib Chemical compound N1=C2N(C3CCCC3)C(=O)C(C(=O)C)=C(C)C2=CN=C1NC(N=C1)=CC=C1N1CCNCC1 AHJRHEGDXFFMBM-UHFFFAOYSA-N 0.000 description 1
- 239000012188 paraffin wax Substances 0.000 description 1
- 238000002161 passivation Methods 0.000 description 1
- 230000001717 pathogenic effect Effects 0.000 description 1
- 229960005547 pelareorep Drugs 0.000 description 1
- 229960005079 pemetrexed Drugs 0.000 description 1
- 239000000137 peptide hydrolase inhibitor Substances 0.000 description 1
- 238000010647 peptide synthesis reaction Methods 0.000 description 1
- 210000001322 periplasm Anatomy 0.000 description 1
- 230000002688 persistence Effects 0.000 description 1
- 229960003742 phenol Drugs 0.000 description 1
- 235000021317 phosphate Nutrition 0.000 description 1
- 150000003013 phosphoric acid derivatives Chemical class 0.000 description 1
- 239000006187 pill Substances 0.000 description 1
- 239000013612 plasmid Substances 0.000 description 1
- 210000004180 plasmocyte Anatomy 0.000 description 1
- 229920001200 poly(ethylene-vinyl acetate) Polymers 0.000 description 1
- 229920000747 poly(lactic acid) Polymers 0.000 description 1
- 229920001223 polyethylene glycol Polymers 0.000 description 1
- 239000004633 polyglycolic acid Substances 0.000 description 1
- 239000004626 polylactic acid Substances 0.000 description 1
- 238000010837 poor prognosis Methods 0.000 description 1
- 230000004481 post-translational protein modification Effects 0.000 description 1
- 230000003389 potentiating effect Effects 0.000 description 1
- 229960000214 pralatrexate Drugs 0.000 description 1
- OGSBUKJUDHAQEA-WMCAAGNKSA-N pralatrexate Chemical compound C1=NC2=NC(N)=NC(N)=C2N=C1CC(CC#C)C1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 OGSBUKJUDHAQEA-WMCAAGNKSA-N 0.000 description 1
- 230000002035 prolonged effect Effects 0.000 description 1
- 238000000159 protein binding assay Methods 0.000 description 1
- 239000008213 purified water Substances 0.000 description 1
- 238000003127 radioimmunoassay Methods 0.000 description 1
- 238000002708 random mutagenesis Methods 0.000 description 1
- 230000010837 receptor-mediated endocytosis Effects 0.000 description 1
- 238000010188 recombinant method Methods 0.000 description 1
- 238000005215 recombination Methods 0.000 description 1
- 230000009467 reduction Effects 0.000 description 1
- 230000002829 reductive effect Effects 0.000 description 1
- 229960004836 regorafenib Drugs 0.000 description 1
- FNHKPVJBJVTLMP-UHFFFAOYSA-N regorafenib Chemical compound C1=NC(C(=O)NC)=CC(OC=2C=C(F)C(NC(=O)NC=3C=C(C(Cl)=CC=3)C(F)(F)F)=CC=2)=C1 FNHKPVJBJVTLMP-UHFFFAOYSA-N 0.000 description 1
- 238000009877 rendering Methods 0.000 description 1
- 229920005989 resin Polymers 0.000 description 1
- 239000011347 resin Substances 0.000 description 1
- 230000004044 response Effects 0.000 description 1
- 230000000979 retarding effect Effects 0.000 description 1
- 238000010839 reverse transcription Methods 0.000 description 1
- 238000004007 reversed phase HPLC Methods 0.000 description 1
- 230000002441 reversible effect Effects 0.000 description 1
- 210000005241 right ventricle Anatomy 0.000 description 1
- 150000003839 salts Chemical class 0.000 description 1
- 230000007017 scission Effects 0.000 description 1
- 239000002356 single layer Substances 0.000 description 1
- 238000002741 site-directed mutagenesis Methods 0.000 description 1
- 229940126586 small molecule drug Drugs 0.000 description 1
- 150000003384 small molecules Chemical class 0.000 description 1
- 230000008410 smoothened signaling pathway Effects 0.000 description 1
- 235000010267 sodium hydrogen sulphite Nutrition 0.000 description 1
- 239000001488 sodium phosphate Substances 0.000 description 1
- 229910000162 sodium phosphate Inorganic materials 0.000 description 1
- 239000007787 solid Substances 0.000 description 1
- 239000002904 solvent Substances 0.000 description 1
- 230000037439 somatic mutation Effects 0.000 description 1
- 229960003787 sorafenib Drugs 0.000 description 1
- 239000000600 sorbitol Substances 0.000 description 1
- 238000001228 spectrum Methods 0.000 description 1
- 238000010186 staining Methods 0.000 description 1
- 210000000130 stem cell Anatomy 0.000 description 1
- 239000008223 sterile water Substances 0.000 description 1
- 238000003860 storage Methods 0.000 description 1
- 150000005846 sugar alcohols Polymers 0.000 description 1
- 150000008163 sugars Chemical class 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
- 229940034785 sutent Drugs 0.000 description 1
- 238000003786 synthesis reaction Methods 0.000 description 1
- 230000001839 systemic circulation Effects 0.000 description 1
- 239000003826 tablet Substances 0.000 description 1
- 229960000235 temsirolimus Drugs 0.000 description 1
- QFJCIRLUMZQUOT-UHFFFAOYSA-N temsirolimus Natural products C1CC(O)C(OC)CC1CC(C)C1OC(=O)C2CCCCN2C(=O)C(=O)C(O)(O2)C(C)CCC2CC(OC)C(C)=CC=CC=CC(C)CC(C)C(=O)C(OC)C(O)C(C)=CC(C)C(=O)C1 QFJCIRLUMZQUOT-UHFFFAOYSA-N 0.000 description 1
- 229960002180 tetracycline Drugs 0.000 description 1
- 229930101283 tetracycline Natural products 0.000 description 1
- 235000019364 tetracycline Nutrition 0.000 description 1
- 150000003522 tetracyclines Chemical class 0.000 description 1
- 229940034915 thalomid Drugs 0.000 description 1
- 231100001274 therapeutic index Toxicity 0.000 description 1
- 229940126622 therapeutic monoclonal antibody Drugs 0.000 description 1
- RTKIYNMVFMVABJ-UHFFFAOYSA-L thimerosal Chemical compound [Na+].CC[Hg]SC1=CC=CC=C1C([O-])=O RTKIYNMVFMVABJ-UHFFFAOYSA-L 0.000 description 1
- 229940033663 thimerosal Drugs 0.000 description 1
- 238000004448 titration Methods 0.000 description 1
- 229960000977 trabectedin Drugs 0.000 description 1
- PKVRCIRHQMSYJX-AIFWHQITSA-N trabectedin Chemical compound C([C@@]1(C(OC2)=O)NCCC3=C1C=C(C(=C3)O)OC)S[C@@H]1C3=C(OC(C)=O)C(C)=C4OCOC4=C3[C@H]2N2[C@@H](O)[C@H](CC=3C4=C(O)C(OC)=C(C)C=3)N(C)[C@H]4[C@@H]21 PKVRCIRHQMSYJX-AIFWHQITSA-N 0.000 description 1
- 238000013518 transcription Methods 0.000 description 1
- 230000035897 transcription Effects 0.000 description 1
- 230000005026 transcription initiation Effects 0.000 description 1
- 230000005030 transcription termination Effects 0.000 description 1
- 230000002103 transcriptional effect Effects 0.000 description 1
- 230000009466 transformation Effects 0.000 description 1
- 108091007466 transmembrane glycoproteins Proteins 0.000 description 1
- 230000032258 transport Effects 0.000 description 1
- 229950001210 trebananib Drugs 0.000 description 1
- 108010075758 trebananib Proteins 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
- RYFMWSXOAZQYPI-UHFFFAOYSA-K trisodium phosphate Chemical compound [Na+].[Na+].[Na+].[O-]P([O-])([O-])=O RYFMWSXOAZQYPI-UHFFFAOYSA-K 0.000 description 1
- 230000004614 tumor growth Effects 0.000 description 1
- 238000004704 ultra performance liquid chromatography Methods 0.000 description 1
- 241001430294 unidentified retrovirus Species 0.000 description 1
- 238000011144 upstream manufacturing Methods 0.000 description 1
- 229960005486 vaccine Drugs 0.000 description 1
- 208000007089 vaccinia Diseases 0.000 description 1
- 210000003462 vein Anatomy 0.000 description 1
- 229960003048 vinblastine Drugs 0.000 description 1
- JXLYSJRDGCGARV-XQKSVPLYSA-N vincaleukoblastine Chemical compound C([C@@H](C[C@]1(C(=O)OC)C=2C(=CC3=C([C@]45[C@H]([C@@]([C@H](OC(C)=O)[C@]6(CC)C=CCN([C@H]56)CC4)(O)C(=O)OC)N3C)C=2)OC)C[C@@](C2)(O)CC)N2CCC2=C1NC1=CC=CC=C21 JXLYSJRDGCGARV-XQKSVPLYSA-N 0.000 description 1
- NMDYYWFGPIMTKO-HBVLKOHWSA-N vinflunine Chemical compound C([C@@](C1=C(C2=CC=CC=C2N1)C1)(C2=C(OC)C=C3N(C)[C@@H]4[C@@]5(C3=C2)CCN2CC=C[C@]([C@@H]52)([C@H]([C@]4(O)C(=O)OC)OC(C)=O)CC)C(=O)OC)[C@H]2C[C@@H](C(C)(F)F)CN1C2 NMDYYWFGPIMTKO-HBVLKOHWSA-N 0.000 description 1
- 229960000922 vinflunine Drugs 0.000 description 1
- 239000013603 viral vector Substances 0.000 description 1
- 230000000007 visual effect Effects 0.000 description 1
- 229940069559 votrient Drugs 0.000 description 1
- 239000003643 water by type Substances 0.000 description 1
- 229940052129 zykadia Drugs 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/50—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
- A61K47/51—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
- A61K47/62—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
- A61K47/65—Peptidic linkers, binders or spacers, e.g. peptidic enzyme-labile linkers
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/50—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
- A61K47/51—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
- A61K47/68—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
- A61K47/6801—Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
- A61K47/6803—Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
- A61K47/68037—Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates the drug being a camptothecin [CPT] or derivatives
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/50—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
- A61K47/51—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
- A61K47/68—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
- A61K47/6835—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
- A61K47/6849—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a receptor, a cell surface antigen or a cell surface determinant
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/50—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
- A61K47/51—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
- A61K47/68—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
- A61K47/6835—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
- A61K47/6851—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/50—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
- A61K47/51—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
- A61K47/68—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
- A61K47/6889—Conjugates wherein the antibody being the modifying agent and wherein the linker, binder or spacer confers particular properties to the conjugates, e.g. peptidic enzyme-labile linkers or acid-labile linkers, providing for an acid-labile immuno conjugate wherein the drug may be released from its antibody conjugated part in an acidic, e.g. tumoural or environment
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
- A61P35/02—Antineoplastic agents specific for leukemia
-
- 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
- C07K16/2881—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against CD71
-
- 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
- C07K2317/33—Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/50—Immunoglobulins specific features characterized by immunoglobulin fragments
- C07K2317/52—Constant or Fc region; Isotype
- C07K2317/524—CH2 domain
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/50—Immunoglobulins specific features characterized by immunoglobulin fragments
- C07K2317/56—Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
- C07K2317/569—Single domain, e.g. dAb, sdAb, VHH, VNAR or nanobody®
-
- 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
- C07K2317/71—Decreased effector function due to an Fc-modification
-
- 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
- C07K2317/77—Internalization into the cell
-
- 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
-
- 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
- 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
- C07K2317/94—Stability, e.g. half-life, pH, temperature or enzyme-resistance
Definitions
- This disclosure relates to anti-TFR1 (transferrin receptor 1) antibodies, antigen-binding fragments, and the uses thereof.
- TFR1 transferrin receptor 1
- Therapeutic antibodies are one of the fastest growing classes of therapeutic compounds, rapidly outpacing the growth of small-molecule drugs. For example, monoclonal antibodies have revolutionized cancer therapy. However, delivery to tumor cells in vivo is hampered by the large size of conventional antibodies.
- the minimal target recognition module of a conventional antibody is composed of two non-covalently associated variable domains (VH and VL) . The inherent hydrophobic interaction of VH and VL domains limits the stability and solubility of engineered antibodies, often causing aggregation and/or mispairing of V-domains.
- VHH variable domain
- Recombinant VHHs are small (15-20 kDa) and strictly monomeric; they bind their target with nM affinity as well as with being stable in a broad pH and temperature ranges. Molecular manipulation is also easier with VHH; this facilitates the production of multivalent formats of monoclonal antibodies compared with conventional recombinant antibodies and their fragments, which is problematic due to aggregation and reduced affinity. Moreover, VHH often binds to epitopes that are less immunogenic for conventional antibodies.
- the therapeutic antibodies are human or humanized antibodies.
- the human or humanized antibodies can be generated by humanization of a rodent antibody (e.g., a mouse antibody) or by using phage libraries.
- rodent antibody e.g., a mouse antibody
- phage libraries usually cannot produce heavy chain antibodies.
- heavy chain antibodies are often obtained from camelid heavy chain antibodies.
- camelid heavy chain antibodies need to be humanized.
- the humanization process may adversely affect the binding affinity and introduce immunogenic epitopes to the antibodies. Iterative and time-consuming experiments are often required to improve the properties of these antibodies. And in some cases, these antibodies can also be immunogenic in patients, leading to attenuation of their efficacy over time. Therefore, there is a need to develop more forms of antibodies to treat or prevent human diseases.
- the present disclosure relates to an antibody or antigen-binding fragment thereof that binds to transferrin receptor 1 (TFR1) , comprising: a heavy chain single variable domain (VHH) comprising complementarity determining regions (CDRs) 1, 2, and 3, in some embodiments, the VHH CDR1 region comprises an amino acid sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identical to a selected VHH CDR1 amino acid sequence, the VHH CDR2 region comprises an amino acid sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identical to a selected VHH CDR2 amino acid sequence, and the VHH CDR3 region comprises an amino acid sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identical to a selected VHH CDR3 amino acid sequence; in some embodiments, the selected VHH CDRs 1, 2, and 3 amino
- VHH CDRs 1, 2, 3 amino acid sequences are set forth in SEQ ID NOs: 1, 2, and 3, respectively;
- VHH CDRs 1, 2, 3 amino acid sequences are set forth in SEQ ID NOs: 4, 5, and 6, respectively;
- VHH CDRs 1, 2, 3 amino acid sequences are set forth in SEQ ID NOs: 7, 8, and 9, respectively;
- VHH CDRs 1, 2, 3 amino acid sequences are set forth in SEQ ID NOs: 10, 11, and 12, respectively;
- VHH CDRs 1, 2, 3 amino acid sequences are set forth in SEQ ID NOs: 13, 14, and 15, respectively;
- VHH CDRs 1, 2, 3 amino acid sequences are set forth in SEQ ID NOs: 16, 17, and 18, respectively;
- VHH CDRs 1, 2, 3 amino acid sequences are set forth in SEQ ID NOs: 19, 20, and 21, respectively;
- VHH CDRs 1, 2, 3 amino acid sequences are set forth in SEQ ID NOs: 22, 23, and 24, respectively.
- the VHH comprises CDRs 1, 2, 3 with the amino acid sequences set forth in SEQ ID NOs: 1, 2, and 3, respectively. In some embodiments, the VHH comprises CDRs 1, 2, 3 with the amino acid sequences set forth in SEQ ID NOs: 4, 5, and 6, respectively. In some embodiments, the VHH comprises CDRs 1, 2, 3 with the amino acid sequences set forth in SEQ ID NOs: 7, 8, and 9, respectively. In some embodiments, the VHH comprises CDRs 1, 2, 3 with the amino acid sequences set forth in SEQ ID NOs: 10, 11, and 12, respectively.
- the disclosure is related to an antibody or antigen-binding fragment thereof that binds to TFR1 comprising a heavy chain single variable region (VHH) comprising an amino acid sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identical to a selected VHH sequence
- the selected VHH sequence is selected from the group consisting of SEQ ID NOs: 25, 26, 27, and 28.
- the VHH comprises the sequence of SEQ ID NO: 25.
- the VHH comprises the sequence of SEQ ID NO: 26.
- the VHH comprises the sequence of SEQ ID NO: 27.
- the VHH comprises the sequence of SEQ ID NO: 28.
- the antibody or antigen-binding fragment specifically binds to a human TFR1, a monkey TFR1, a mouse TFR1, or a chimeric TFR1.
- the antibody or antigen-binding fragment is a human or humanized antibody or antigen-binding fragment thereof.
- the antibody or antigen-binding fragment is a multi-specific antibody (e.g., a bispecific antibody) .
- the disclosure is related to an antibody or antigen-binding fragment thereof comprising the VHH CDRs 1, 2, 3, of the antibody or antigen-binding fragment thereof as described herein.
- the antibody or antigen-binding fragment comprises a human IgG Fc (e.g., a human IgG1 Fc) .
- the human IgG Fc comprises a non-asparagine residue (e.g., alanine) at position 297 according to EU numbering.
- the antibody or antigen-binding fragment comprises two or more heavy chain single variable domains.
- the disclosure is related to a nucleic acid comprising a polynucleotide encoding the antibody or antigen-binding fragment thereof as described herein.
- the nucleic acid is cDNA.
- the disclosure is related to a vector comprising one or more of the nucleic acids as described herein.
- the disclosure is related to a cell comprising the vector as described herein.
- the cell is a CHO cell.
- the disclosure is related to a cell comprising one or more of the nucleic acids described herein.
- the disclosure is related to a method of producing an antibody or an antigen-binding fragment thereof, the method comprising (a) culturing the cell as described herein under conditions sufficient for the cell to produce the antibody or the antigen-binding fragment thereof; and (b) collecting the antibody or the antigen-binding fragment thereof produced by the cell.
- the disclosure is related to an antibody-drug conjugate comprising the antibody or antigen-binding fragment thereof as described herein covalently bound to a therapeutic agent.
- the therapeutic agent is a cytotoxic or cytostatic agent.
- the disclosure is related to a method of treating a subject having a brain disease (e.g., a brain cancer) , the method comprising administering a therapeutically effective amount of a composition comprising the antibody or antigen-binding fragment thereof, or the antibody-drug conjugate as described herein, to the subject.
- a brain disease e.g., a brain cancer
- the antibody or antigen-binding fragment thereof, or the antibody-drug conjugate can pass across the blood-brain barrier (BBB) of the subject.
- BBB blood-brain barrier
- the disclosure is related to a method of treating a subject having a cancer, the method comprising administering a therapeutically effective amount of a composition comprising the antibody or antigen-binding fragment thereof, or the antibody-drug conjugate as described herein, to the subject.
- the cancer is brain cancer, lung cancer, gastric cancer, colorectal cancer, liver cancer, ovarian cancer, prostate cancer, leukemia, or breast cancer.
- the disclosure is related to a method of identifying a subject as having a brain disease (e.g., a brain cancer) , the method comprising detecting a sample collected from the subject as having the brain disease by the antibody or antigen-binding fragment thereof as described herein, thereby identifying the subject as having the brain disease.
- the sample is a brain parenchyma sample from the subject.
- the subject described herein is a human subject.
- the disclosure is related to a method of delivering an agent to cross blood brain barrier, the method comprising administering the agent covalently linked to the antibody or antigen-binding fragment thereof as described herein to the subject.
- the agent is an antibody or an antibody drug conjugate.
- the agent is anti-amyloid antibody.
- the disclosure is related to a pharmaceutical composition comprising the antibody or antigen-binding fragment thereof as described herein, and a pharmaceutically acceptable carrier. In one aspect, the disclosure is related to a pharmaceutical composition comprising the antibody drug conjugate as described herein, and a pharmaceutically acceptable carrier.
- the disclosure is related to an antibody or antigen-binding fragment thereof that cross-competes with the antibody or antigen-binding fragment thereof as described herein.
- the disclosure provides a method of making an antibody that specifically binds to an antigen.
- the method involves exposing the animal as described herein to the antigen; obtaining the sequence of (e.g. by sequencing) nucleic acids encoding human heavy chain immunoglobulin variable regions in a cell that expresses a chimeric heavy chain antibody that specifically binds to the antigen; and operably linking in a cell the nucleic acid encoding the human heavy chain immunoglobulin variable region with a nucleic acid encoding a human heavy chain immunoglobulin constant region.
- the disclosure also relates to an offspring of the non-human mammal.
- the non-human mammal is a rodent.
- the non-human mammal is a mouse.
- the disclosure also provides to a cell including the targeting vector as described herein.
- the disclosure also relates to a cell (e.g., a stem cell, an embryonic stem cell, an immune cell, a B cell, a T cell, or a hybridoma) or a cell line, or a primary cell culture thereof derived from the non-human mammal or an offspring thereof.
- the disclosure further relates to the tissue, organ or a culture thereof derived from the non-human mammal or an offspring thereof.
- the disclosure further relates to the use of the non-human mammal or an offspring thereof, the animal model generated through the method as described herein in the development of a product related to an immunization process, the manufacture of a human antibody, or the model system for research in pharmacology, immunology, microbiology and medicine.
- FIG. 1 lists CDR sequences of heavy chain variable region of anti-TFR1 antibodies according to Kabat numbering.
- FIG. 2 lists CDR sequences of heavy chain variable region of anti-TFR1 antibodies according to IMGT numbering.
- FIG. 3 lists amino acids sequences discussed in the disclosure.
- FIG. 4A shows the antibody concentration in total brain protein of hTFR1 mice within 72 hours of intravenous (i. v. ) administration of hIgG1 (G1) , JR141-N (G2) , 23B8-N (G3) , 24A1-N (G4) , 24G5-N (G5) , or 24C9-N (G6) .
- FIG. 4B shows the ratio of antibody concentration in brain total protein to serum antibody concentration of hTFR1 mice within 72 hours of intravenous (i. v. ) administration of hIgG1 (G1) , JR141-N (G2) , 23B8-N (G3) , 24A1-N (G4) , 24G5-N (G5) , or 24C9-N (G6) .
- FIG. 4C shows the antibody concentration in brain parenchyma of hTFR1 mice within 72 hours of intravenous (i. v. ) administration of hIgG1 (G1) , JR141-N (G2) , 23B8-N (G3) , 24A1-N (G4) , 24G5-N (G5) , or 24C9-N (G6) .
- FIG. 4D shows the ratio of antibody concentration in brain parenchyma to serum antibody concentration of hTFR1 mice within 72 hours of intravenous (i. v. ) administration of hIgG1 (G1) , JR141-N (G2) , 23B8-N (G3) , 24A1-N (G4) , 24G5-N (G5) , or 24C9-N (G6) .
- FIG. 5A shows the antibody concentration test results in brain parenchyma after 24 hours of intravenous (i. v. ) administration of hIgG1 (G1) , JR141-N (G2) , 23B8-N (G3) , 24A1-N (G4) , or 24G5-N (G5) .
- FIG. 5B shows the antibody concentration test results in brain total protein (Whole Brain) after 24 hours of intravenous (i. v. ) administration of hIgG1 (G1) , JR141-N (G2) , 23B8-N (G3) , 24A1-N (G4) , or 24G5-N (G5) .
- FIG. 6 shows the antibody concentration results after 6 hours or 24 hours of intravenous (i.v. ) administration of JR141-N (G2-G4) or 24G5-N (G5-G7) ) .
- hIgG1 was used as a negative control.
- FIG. 7 shows the ADC concentration test results in brain parenchyma after 72 hours of intravenous (i. v. ) administration of 24G5-ADC or 24G5-mono-ADC.
- a heavy-chain antibody is an antibody which has only heavy chains (generally two heavy chains) and lacks the two light chains usually found in antibodies.
- Naturally occurring heavy-chain antibodies have been discovered in cartilaginous fishes (e.g., shark) and camelids (e.g., llama) .
- the immunoglobulin new antigen receptor IgNAR
- IgNAR shows significant structural differences to other antibodies. It has five constant domains (CH) per chain instead of the usual three, several disulfide bonds in unusual positions, and the complementarity-determining region 3 (CDR3) forms an extended loop covering the site which binds to a light chain in other antibodies.
- camelids such as dromedaries, camels, llamas and alpacas.
- camelids e.g., llamas
- camelids can produce conventional antibodies made of two heavy chains and two light chains bound together with disulfide bonds in a Y shape (e.g., IgG1) .
- IgG1 two unique subclasses of IgG: IgG2 and IgG3, also known as heavy chain IgG.
- IgG2 and IgG3 also known as heavy chain IgG.
- These antibodies are made of only two heavy chains, which lack the CH1 region but still bear an antigen-binding domain (e.g., VHH) at their N-terminus.
- an antigen-binding domain e.g., VHH
- variable regions from both heavy and light chains to allow a high diversity of antigen-antibody interactions. Although isolated heavy and light chains still show this capacity, they exhibit very low affinity when compared to paired heavy and light chains.
- the unique feature of heavy chain IgG is the capacity of their monomeric antigen binding regions to bind antigens with specificity, affinity and especially diversity that are comparable to conventional antibodies without the need of pairing with another region. This feature is mainly due to a couple of major variations within the amino acid sequence of the variable region of the two heavy chains, which induce deep conformational changes when compared to conventional Ig. Major substitutions in the variable regions prevent the light chains from binding to the heavy chains, but also prevent unbound heavy chains from being recycled by the Immunoglobulin Binding Protein.
- VHH single variable domain of these heavy-chain antibodies
- sdAb single variable domain generated by adaptive immune systems.
- CDR3 Complementarity Determining Region 3
- VHHs can extend into crevices on proteins that are not accessible to conventional antibodies, including functionally interesting sites such as the active site of an enzyme or the receptor-binding canyon on a virus surface.
- an additional cysteine residue allows the structure to be more stable, thus increasing the strength of the interaction.
- VHHs offer numerous other advantages compared to conventional antibodies carrying variable domains (VH and VL) of conventional antibodies, including higher stability, solubility, expression yields, and refolding capacity, as well as better in vivo tissue penetration. Moreover, in contrast to the VH domains of conventional antibodies, VHH do not display an intrinsic tendency to bind to light chains. This facilitates the induction of heavy chain antibodies in the presence of a functional light chain loci. Further, since VHH do not bind to VL domains, it is much easier to reformat VHHs into bispecific antibody constructs than constructs containing conventional VH-VL pairs or single domains based on VH domains.
- the CDR3 corresponds to the unique region of the antibody molecule that is encoded by a DNA element newly generated during B-cell development. Genetic recombination results in the fusion of a D-element with flanking V-and J-elements. During recombination further genetic diversity is generated by addition and/or deletion of nucleotides at the junctions. Thereby, the CDR3 loop provides the major contribution to antibody diversity and specificity.
- variable region genes IGHV, IGHD, and IGHJ
- IGHV variable region genes
- antibody refers to any antigen-binding molecule that contains at least one (e.g., one, two, three, four, five, or six) complementary determining region (CDR) (e.g., any of the three CDRs from an immunoglobulin light chain or any of the three CDRs from an immunoglobulin heavy chain) and is capable of specifically binding to an epitope.
- CDR complementary determining region
- Non-limiting examples of antibodies include: monoclonal antibodies, polyclonal antibodies, multi-specific antibodies (e.g., bi-specific antibodies) , single-chain antibodies, heavy chain antibodies, chimeric antibodies, human antibodies, and humanized antibodies.
- an antibody can contain an Fc region of a human antibody.
- the term antibody also includes derivatives, e.g., bi-specific antibodies, single-chain antibodies, diabodies, linear antibodies, and multi-specific antibodies formed from antibody fragments.
- the term “antigen-binding fragment” refers to a portion of a full-length antibody, wherein the portion is capable of specifically binding to an antigen.
- the antigen-binding fragment contains at least one variable domain (e.g., a variable domain of a heavy chain or a variable domain of light chain) .
- variable domains include, e.g., Fab, Fab’, F (ab’) 2, and Fv fragments.
- human antibody refers to an antibody that is encoded by a nucleic acid (e.g., rearranged human immunoglobulin heavy or light chain locus) present in a human.
- a human antibody is collected from a human or produced in a human cell culture (e.g., human hybridoma cells) .
- a human antibody is produced in a non-human cell (e.g., a mouse or hamster cell line) .
- a human antibody is produced in a bacterial or yeast cell.
- a human antibody is produced in a transgenic non-human animal (e.g., a mouse) containing an unrearranged or rearranged human immunoglobulin locus (e.g., heavy or light chain human immunoglobulin locus) .
- a transgenic non-human animal e.g., a mouse
- an unrearranged or rearranged human immunoglobulin locus e.g., heavy or light chain human immunoglobulin locus
- chimeric antibody refers to an antibody that contains a sequence present in at least two different antibodies (e.g., antibodies from two different mammalian species such as a human and a mouse antibody) .
- a non-limiting example of a chimeric antibody is an antibody containing the variable domain sequences (e.g., all or part of a light chain and/or heavy chain variable domain sequence) of a human antibody and the constant domains of a non-human antibody. Additional examples of chimeric antibodies are described herein and are known in the art.
- humanized antibody refers to a non-human antibody which contains sequence derived from a non-human (e.g., mouse) immunoglobulin and contains sequences derived from a human immunoglobulin.
- single-chain antibody refers to a single polypeptide that contains at least two immunoglobulin variable domains (e.g., a variable domain of a mammalian immunoglobulin heavy chain or light chain) that is capable of specifically binding to an antigen.
- heavy-chain antibody refers to an antibody molecule which is composed only of heavy chains (generally two) and does not have any light chains.
- VHH refers to the variable domain derived from a heavy-chain antibody.
- the VHH can specifically recognize an antigen without the need to be paired with a VL.
- the VHH (also know as sdAb or nanobody) described herein is derived from any of the humanized heavy-chain antibody described herein.
- the VHH, sdAb, or nanobody described herein is derived from the heavy chain antibody produced by any of the genetically modified non-human animal described herein.
- the terms “subject” and “patient” are used interchangeably throughout the specification and describe an animal, human or non-human. Veterinary and non-veterinary applications are contemplated by the present disclosure.
- Human patients can be adult humans or juvenile humans (e.g., humans below the age of 18 years old) .
- patients include but are not limited to mice, rats, hamsters, guinea-pigs, rabbits, ferrets, cats, dogs, and primates.
- non-human primates e.g., monkey, chimpanzee, gorilla, and the like
- rodents e.g., rats, mice, gerbils, hamsters, ferrets, rabbits
- lagomorphs e.g., swine (e.g., pig, miniature pig)
- equine canine, feline, bovine, and other domestic, farm, and zoo animals.
- the phrases “specifically binding” and “specifically binds” mean that the antibody interacts with its target molecule preferably to other molecules, because the interaction is dependent upon the presence of a particular structure (i.e., the antigenic determinant or epitope) on the target molecule; in other words, the reagent is recognizing and binding to molecules that include a specific structure rather than to all molecules in general.
- An antibody that specifically binds to the target molecule may be referred to as a target-specific antibody.
- polypeptide, ” “peptide, ” and “protein” are used interchangeably to refer to polymers of amino acids of any length of at least two amino acids.
- nucleic acid molecule As used herein, the terms “polynucleotide, ” “nucleic acid molecule, ” and “nucleic acid sequence” are used interchangeably herein to refer to polymers of nucleotides of any length of at least two nucleotides, and include, without limitation, DNA, RNA, DNA/RNA hybrids, and modifications thereof.
- the present disclosure provides antibodies and antigen-binding fragments thereof (e.g., heavy chain antibodies, humanized heavy chain antibodies, or multi-specific antibodies) that are produced by the methods described herein.
- antibodies and antigen-binding fragments thereof e.g., heavy chain antibodies, humanized heavy chain antibodies, or multi-specific antibodies
- a non-limiting antibody of the present disclosure can be an intact, four-immunoglobulin-chain antibody comprising two heavy chains and two light chains.
- the heavy chain of the antibody can be of any isotype including IgM, IgG, IgE, IgA, or IgD or subclasses including IgG1, IgG2, IgG2a, IgG2b, IgG3, IgG4, IgE1, IgE2, etc.
- the light chain can be a kappa light chain or a lambda light chain.
- An antibody can comprise two identical copies of a light chain and two identical copies of a heavy chain.
- the heavy chains which each contain one variable domain (or variable region, V H ) and multiple constant domains (or constant regions) , bind to one another via disulfide bonding within their constant domains to form the “stem” of the antibody.
- the light chains which each contain one variable domain (or variable region, V L ) and one constant domain (or constant region) , each bind to one heavy chain via disulfide binding.
- the variable region of each light chain is aligned with the variable region of the heavy chain to which it is bound.
- the variable regions of both the light chains and heavy chains contain three hypervariable regions sandwiched between more conserved framework regions (FR) .
- CDRs complementary determining regions
- the four framework regions largely adopt a beta-sheet conformation and the CDRs form loops connecting, and in some cases forming part of, the beta-sheet structure.
- the CDRs in each chain are held in close proximity by the framework regions and, with the CDRs from the other chain, contribute to the formation of the antigen-binding region.
- the CDRs are important for recognizing an epitope of an antigen.
- an “epitope” is the smallest portion of a target molecule capable of being specifically bound by the antigen binding domain of an antibody.
- the minimal size of an epitope may be about three, four, five, six, or seven amino acids, but these amino acids need not be in a consecutive linear sequence of the antigen’s primary structure, as the epitope may depend on an antigen’s three-dimensional configuration based on the antigen’s secondary and tertiary structure.
- the antibody is an intact immunoglobulin molecule (e.g., IgG1, IgG2a, IgG2b, IgG2c, IgG3, IgG4, IgM, IgD, IgE, IgA) .
- the IgG subclasses (IgG1, IgG2, IgG3, and IgG4) are highly conserved, differ in their constant region, particularly in their hinges and upper CH2 domains. The sequences and differences of the IgG subclasses are known in the art, and are described, e.g., in Vidarsson, et al, "IgG subclasses and allotypes: from structure to effector functions.
- the heavy chain constant regions in the heavy chain antibodies can be derived from any immunoglobulin molecules described herein (e.g., IgG1, IgG2a, IgG2b, IgG2c, IgG3, IgG4, IgM, IgD, IgE, IgA) .
- the antibody can also be an immunoglobulin molecule that is derived from any species (e.g., human, rodent, mouse, rat, camelid) .
- Antibodies disclosed herein also include, but are not limited to, polyclonal, monoclonal, monospecific, multi-specific antibodies, and chimeric antibodies that include an immunoglobulin binding domain fused to another polypeptide.
- the term “antigen binding domain” or “antigen binding fragment” is a portion of an antibody that retains specific binding activity of the intact antibody, i.e., any portion of an antibody that is capable of specific binding to an epitope on the intact antibody’s target molecule.
- an antibody or an antigen binding fragment thereof can be, e.g., a scFv, a Fv, a Fd, a dAb, a bispecific antibody, a bispecific scFv, a diabody, a linear antibody, a single-chain antibody molecule, a multi-specific antibody formed from antibody fragments, and any polypeptide that includes a binding domain which is, or is homologous to, an antibody binding domain.
- Non-limiting examples of antigen binding domains include, e.g., the heavy chain and/or light chain CDRs of an intact antibody, the heavy and/or light chain variable regions of an intact antibody, full length heavy or light chains of an intact antibody, or an individual CDR from either the heavy chain or the light chain of an intact antibody.
- the antigen binding fragment can form a part of a chimeric antigen receptor (CAR) .
- the chimeric antigen receptor are fusions of VHH as described herein, fused to CD3-zeta transmembrane-and endodomain.
- the antibodies and antigen-binding fragments thereof (e.g., humanized antibodies or chimeric antibodies) that are produced by the methods described herein have various advantages. In some embodiments, no further optimization is required to obtain desired properties (e.g., binding affinities, thermal stabilities, and/or limited aggregation) .
- the antibody (or antigen-binding fragments thereof) specifically binds to a target with a dissociation rate (koff) of less than 0.1 s -1 , less than 0.01 s -1 , less than 0.001 s -1 , less than 0.0001 s -1 , or less than 0.00001 s -1 .
- the dissociation rate (koff) is greater than 0.01 s -1 , greater than 0.001 s -1 , greater than 0.0001 s -1 , greater than 0.00001 s -1 , or greater than 0.000001 s -1 .
- kinetic association rates are greater than 1 x 10 2 /Ms, greater than 1 x 10 3 /Ms, greater than 1 x 10 4 /Ms, greater than 1 x 10 5 /Ms, or greater than 1 x 10 6 /Ms. In some embodiments, kinetic association rates (kon) are less than 1 x 10 5 /Ms, less than 1 x 10 6 /Ms, or less than 1 x 10 7 /Ms.
- KD is less than 1 x 10 -6 M, less than 1 x 10 -7 M, less than 1 x 10 -8 M, less than 1 x 10 -9 M, or less than 1 x 10 -10 M. In some embodiments, the KD is less than 50 nM, 40 nM, 30 nM, 20 nM, 15 nM, 10 nM, 9 nM, 8 nM, 7 nM, 6 nM, 5 nM, 4 nM, 3 nM, 2 nM, or 1 nM.
- KD is greater than 1 x 10 -7 M, greater than 1 x 10 -8 M, greater than 1 x 10 - 9 M, greater than 1 x 10 -10 M, greater than 1 x 10 -11 M, or greater than 1 x 10 -12 M.
- the antibody binds to a target with KD less than or equal to about 0.9 nM, 0.8 nM, 0.7 nM, 0.6 nM, 0.5 nM, 0.4 nM, 0.3 nM, 0.2 nM, or 0.1 nM.
- thermal stabilities are determined.
- the antibodies or antigen binding fragments as described herein can have a Tm greater than 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, or 95 °C.
- substitutions are performed to a parental heavy chain antibody sequence to make a variant heavy chain antibody.
- a heavy chain antibody variant of a parental heavy chain antibody has an antigen binding affinity that is at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%or at least 100% (e.g., at least 150%, at least 200%, at least 500%, at least 1000%, or up to at least 10,000%) of the binding affinity of the parental heavy chain antibody to a particular antigen.
- a variant heavy chain antibody will comprise a single substitution as compared to a parental heavy chain antibody.
- the resultant variant heavy chain antibody is tested to confirm that the desired binding affinity and/or specificity has not been significantly decreased by the replacement residues.
- an improved variant heavy chain antibody is produced by the substitution of amino acids from a different human heavy chain sequence.
- the VHH is at least 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%identical to a parental VHH.
- the VHH described herein can be used to make multi-specific (e.g., bispecific antibodies) .
- the present disclosure provides a multi-specific antibody comprising: a first antigen binding portion and a second antigen binding portion.
- the first antigen binding portion comprises a heavy chain variable domain (VH) and a light chain variable domain (VL) , wherein the VH and VL together form an antigen-binding site that specifically binds a first epitope.
- the first antigen binding portion comprises a VHH that specifically binds a first epitope.
- the second antigen binding portion comprises a VHH that specifically binds a second epitope.
- the first epitope and the second epitope are from the same antigen.
- the first epitope and the second epitope are from different antigens.
- the first antigen binding portion is a full-length antibody consisting of two heavy chains and two light chains. In some embodiments, the first antigen binding portion is an antibody fragment comprising a heavy chain comprising the VH and a light chain comprising the VL. In some embodiments, the second antigen binding portion comprises a single polypeptide chain. In some embodiments, the C terminus of the second antigen binding portion is fused to the N-terminus of at least one heavy chain of the first antigen binding portion. In some embodiments, the C terminus of the second antigen binding portion is fused to the N-terminus of at least one light chain of the first antigen binding portion.
- the N terminus of the second antigen binding portion is fused to the C-terminus of at least one heavy chain of the first antigen binding portion. In some embodiments, the N terminus of the second antigen binding portion is fused to the C-terminus of at least one light chain of the first antigen binding portion. In some embodiments, the second antigen binding portion is a Fab-like domain comprising a first polypeptide chain comprising a first VHH fused to a CH1 domain, and a second polypeptide chain comprising a second VHH fused to a CL domain.
- the antibody or antigen-binding fragment thereof is a tri-specific antibody.
- the tri-specific antibody is a tri-specific VHH-Fc.
- the tri-specific antibody comprises the same VHHs.
- the tri-specific antibody comprises different VHHs.
- the VHHs bind to the same epitope. In some embodiments, the VHHs bind to different epitopes.
- the antibody or antigen-binding fragment thereof has four or more than four VHHs.
- at least four VHHs are combined without the addition of IgG Fc domain to construct tetra-specific VHHs. These molecules would have the added advantage of increased affinity and avidity towards the antigen compared to bi-and tri-specific VHH-Fcs, despite lacking the Fc effector functions.
- these the antibody or antigen-binding fragment thereof (e.g., comprising 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more than 10) has a functional Fc.
- the heavy-chain antibody produced by the genetically modified non-human animal described herein has a VHH domain that includes CDR1, CDR2, and CDR3.
- the CDR3 length is between 6-23, e.g., 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22 or 23.
- the CDR3 length is at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, or at least 23.
- TFR1 Transferrin receptor 1 (TFR1)
- TFR1 also known as cluster of differentiation 71 (CD71)
- CD71 cluster of differentiation 71
- Tf transferrin
- Human TFR1 is a 90 kDa type II transmembrane glycoprotein consisting of 760 amino acids that is found as a dimer (180 kDa) linked by disulfide bonds on the cell surface.
- the TFR1 monomer is composed of a large extracellular, C-terminal domain of 671 amino acids containing the Tf-binding site, a transmembrane domain (28 amino acids) , and an intracellular N-terminal domain (61 amino acids) .
- the C-terminal extracellular domain contains three N-linked glycosylation sites at asparagine residues 251, 317, and 727 and one O-linked glycosylation site at threonine 104, which are all required for adequate function of the receptor.
- Transferrin It is an 80 kDa glycoprotein composed of two 40 kDa subunits, known as the N-and C-lobes that are separated by a short linker sequence. Each subunit is capable of binding to one free ferric iron (Fe 3+ ) and thus, Tf may have up to two atoms of iron attached. Tf in its iron free form, apo-Tf, binds Fe 3+ with high efficiency in the blood and transports it to the cell surface for internalization through the interaction with TFR1. As a membrane protein regulating iron import, TFR1 is a member of the TFR family that shows nanomolar affinity to transferrin (Tf) bound to Fe 3+ .
- Tf-TFR1 The complex of Tf-TFR1 is internalized through endocytosis mediated by clathrin, and Fe 3+ is disassociated from Tf when pH decreases to 5.5. At this pH, apo-Tf and TFR1 are still associated and recycled to cell surface with physiological pH, so the former is released.
- TFR1 Iron uptake by transferrin receptor is an important way for cancer cells to absorb iron, thus accumulating evidence has proven that TFR1 participated in tumor onset and progression, and its expression was dysregulated significantly in many cancers. The relationship between TFR1 and cancers has been revealed, rendering TFR1 a valuable pharmaceutical target for intervening with cancers.
- TFR1 expressed on the endothelial cells of the blood-brain barrier is used also in preclinical research to allow the delivery of large molecules including antibodies into the brain.
- the TFR1 targeting antibodies can cross the blood-brain barrier, without interfering with the uptake of iron.
- TFR1, Tf, and their functions can be found, e.g., in Candelaria, P.V., et al. "Antibodies targeting the transferrin receptor 1 (TfR1) as direct anti-cancer agents. " Frontiers in Immunology 12 (2021) : 607692; and Shen, Y., et al. "Transferrin receptor 1 in cancer: a new sight for cancer therapy. " American Journal of Cancer Research 8.6 (2018) : 916; each of which is incorporated by reference in its entirety.
- VHH Heavy chain single variable domain
- Monoclonal and recombinant antibodies are important tools in medicine and biotechnology. Like all mammals, camelids (e.g., llamas) can produce conventional antibodies made of two heavy chains and two light chains bound together with disulfide bonds in a Y shape (e.g., IgG1) . However, they also produce two unique subclasses of IgG: IgG2 and IgG3, also known as heavy chain IgG. These antibodies are made of only two heavy chains, which lack the CH1 region but still bear an antigen-binding domain at their N-terminus called VHH (or nanobody) . Conventional Ig require the association of variable regions from both heavy and light chains to allow a high diversity of antigen-antibody interactions.
- heavy chain IgG The unique feature of heavy chain IgG is the capacity of their monomeric antigen binding regions to bind antigens with specificity, affinity and especially diversity that are comparable to conventional antibodies without the need of pairing with another region. This feature is mainly due to a couple of major variations within the amino acid sequence of the variable region of the two heavy chains, which induce deep conformational changes when compared to conventional Ig. Major substitutions in the variable regions prevent the light chains from binding to the heavy chains, but also prevent unbound heavy chains from being recycled by the immunoglobulin binding proteins.
- the single variable domain of these antibodies (designated VHH, sdAb, or nanobody) is the smallest antigen-binding domain generated by adaptive immune systems.
- the third Complementarity Determining Region (CDR3) of the variable region of these antibodies has been found to be twice as long as the conventional ones. This results in an increased interaction surface with the antigen as well as an increased diversity of antigen-antibody interactions, which compensates the absence of the light chains.
- CDR3 complementarity-determining region 3
- VHHs can extend into crevices on proteins that are not accessible to conventional antibodies, including functionally interesting sites such as the active site of an enzyme or the receptor-binding canyon on a virus surface.
- VHHs offer numerous other advantages compared to conventional antibodies carrying variable domains (VH and VL) of conventional antibodies, including higher stability, solubility, expression yields, and refolding capacity, as well as better in vivo tissue penetration and internalization. Moreover, in contrast to the VH domains of conventional antibodies, VHH do not display an intrinsic tendency to bind to light chains. Since VHH do not bind to VL domains, it is much easier to reformat VHHs into multi-specific (e.g., bispecific antibody) constructs than constructs containing conventional VH-VL pairs or single domains based on VH domains.
- VH and VL variable domains
- the disclosure provides e.g., anti-TFR1 antibodies, the modified antibodies thereof, the chimeric antibodies thereof, and the humanized antibodies thereof.
- the CDR sequences for 23B8, and 23B8 derived antibodies include CDRs of the VHH domain as set forth in SEQ ID NOs: 1, 2, and 3, respectively, as defined by Kabat numbering.
- the CDRs can also be defined by IMGT system. Under the IMGT numbering, the CDRs of the VHH domain are set forth in SEQ ID NOs: 13, 14, and 15, respectively.
- the CDR sequences for 24A1, and 24A1 derived antibodies include CDRs of the VHH domain as set forth in SEQ ID NOs: 4, 5, and 6, respectively, as defined by Kabat numbering.
- the CDRs can also be defined by IMGT system. Under the IMGT numbering, the CDRs of the VHH domain are set forth in SEQ ID NOs: 16, 17, and 18, respectively.
- the CDR sequences for 24C9, and 24C9 derived antibodies include CDRs of the VHH domain as set forth in SEQ ID NOs: 7, 8, and 9, respectively, as defined by Kabat numbering.
- the CDRs can also be defined by IMGT system. Under the IMGT numbering, the CDRs of the VHH domain are set forth in SEQ ID NOs: 19, 20, and 21, respectively.
- the CDR sequences for 24G5, and 24G5 derived antibodies include CDRs of the VHH domain as set forth in SEQ ID NOs: 10, 11, and 12, respectively, as defined by Kabat numbering.
- the CDRs can also be defined by IMGT system. Under the IMGT numbering, the CDRs of the VHH domain are set forth in SEQ ID NOs: 22, 23, and 24, respectively.
- the amino acid sequence for the VHH domain of 23B8 antibody is set forth in SEQ ID NO: 25.
- the amino acid sequence for the VHH domain of 24A1 antibody is set forth in SEQ ID NO: 26.
- the amino acid sequence for the VHH domain of 24C9 antibody is set forth in SEQ ID NO: 27.
- the amino acid sequence for the VHH domain of 24G5 antibody is set forth in SEQ ID NO: 28.
- the amino acid sequences for various modified or humanized VHH are also provided.
- the VHH domain of a heavy-chain antibody can have more than one version of humanized sequences.
- the humanized VHH domain is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%identical to any sequence of SEQ ID NOS: 25-28.
- the antibodies or antigen-binding fragments thereof described herein can also contain one, two, or three VHH domain CDRs selected from the group of SEQ ID NOs: 1-3, SEQ ID NOs: 4-6, SEQ ID NOs: 7-9, SEQ ID NOs: 10-12, SEQ ID NOs: 13-15, SEQ ID NOs: 16-18, SEQ ID NOs: 19-21, and SEQ ID NOs: 22-24.
- the antibodies can have a heavy chain single variable domain (VHH) comprising complementarity determining regions (CDRs) 1, 2, 3, wherein the CDR1 region comprises or consists of an amino acid sequence that is at least 80%, 85%, 90%, or 95%identical to a selected VHH CDR1 amino acid sequence, the CDR2 region comprises or consists of an amino acid sequence that is at least 80%, 85%, 90%, or 95%identical to a selected VHH CDR2 amino acid sequence, and the CDR3 region comprises or consists of an amino acid sequence that is at least 80%, 85%, 90%, or 95%identical to a selected VHH CDR3 amino acid sequence.
- VHH CDRs 1, 2, 3 amino acid sequences are shown in FIG. 1 and FIG. 2.
- the antibody or an antigen-binding fragment described herein can contain a heavy chain single variable domain (VHH) containing one, two, or three of VHH CDR1 with zero, one or two amino acid insertions, deletions, or substitutions; VHH CDR2 with zero, one or two amino acid insertions, deletions, or substitutions; VHH CDR3 with zero, one or two amino acid insertions, deletions, or substitutions, wherein VHH CDR1, VHH CDR2, and VHH CDR3 are selected from the CDRs in FIG. 3.
- VHH heavy chain single variable domain
- the antibody or an antigen-binding fragment described herein can contain a heavy chain single variable domain (VHH) containing one, two, or three of the CDRs of SEQ ID NO: 1 with zero, one or two amino acid insertions, deletions, or substitutions; SEQ ID NO: 2 with zero, one or two amino acid insertions, deletions, or substitutions; SEQ ID NO: 3 with zero, one or two amino acid insertions, deletions, or substitutions.
- VHH heavy chain single variable domain
- the antibody or an antigen-binding fragment described herein can contain a heavy chain single variable domain (VHH) containing one, two, or three of the CDRs of SEQ ID NO: 4 with zero, one or two amino acid insertions, deletions, or substitutions; SEQ ID NO: 5 with zero, one or two amino acid insertions, deletions, or substitutions; SEQ ID NO: 6 with zero, one or two amino acid insertions, deletions, or substitutions.
- VHH heavy chain single variable domain
- the antibody or an antigen-binding fragment described herein can contain a heavy chain single variable domain (VHH) containing one, two, or three of the CDRs of SEQ ID NO: 7 with zero, one or two amino acid insertions, deletions, or substitutions; SEQ ID NO: 8 with zero, one or two amino acid insertions, deletions, or substitutions; SEQ ID NO: 9 with zero, one or two amino acid insertions, deletions, or substitutions.
- VHH heavy chain single variable domain
- the antibody or an antigen-binding fragment described herein can contain a heavy chain single variable domain (VHH) containing one, two, or three of the CDRs of SEQ ID NO: 10 with zero, one or two amino acid insertions, deletions, or substitutions; SEQ ID NO: 11 with zero, one or two amino acid insertions, deletions, or substitutions; SEQ ID NO: 12 with zero, one or two amino acid insertions, deletions, or substitutions.
- VHH heavy chain single variable domain
- the antibody or an antigen-binding fragment described herein can contain a heavy chain single variable domain (VHH) containing one, two, or three of the CDRs of SEQ ID NO: 13 with zero, one or two amino acid insertions, deletions, or substitutions; SEQ ID NO: 14 with zero, one or two amino acid insertions, deletions, or substitutions; SEQ ID NO: 15 with zero, one or two amino acid insertions, deletions, or substitutions.
- VHH heavy chain single variable domain
- the antibody or an antigen-binding fragment described herein can contain a heavy chain single variable domain (VHH) containing one, two, or three of the CDRs of SEQ ID NO: 16 with zero, one or two amino acid insertions, deletions, or substitutions; SEQ ID NO: 17 with zero, one or two amino acid insertions, deletions, or substitutions; SEQ ID NO: 18 with zero, one or two amino acid insertions, deletions, or substitutions.
- VHH heavy chain single variable domain
- the antibody or an antigen-binding fragment described herein can contain a heavy chain single variable domain (VHH) containing one, two, or three of the CDRs of SEQ ID NO: 19 with zero, one or two amino acid insertions, deletions, or substitutions; SEQ ID NO: 20 with zero, one or two amino acid insertions, deletions, or substitutions; SEQ ID NO: 21 with zero, one or two amino acid insertions, deletions, or substitutions.
- VHH heavy chain single variable domain
- the antibody or an antigen-binding fragment described herein can contain a heavy chain single variable domain (VHH) containing one, two, or three of the CDRs of SEQ ID NO: 22 with zero, one or two amino acid insertions, deletions, or substitutions; SEQ ID NO: 23 with zero, one or two amino acid insertions, deletions, or substitutions; SEQ ID NO: 24 with zero, one or two amino acid insertions, deletions, or substitutions.
- VHH heavy chain single variable domain
- the insertions, deletions, and substitutions can be within the CDR sequence, or at one or both terminal ends of the CDR sequence.
- the CDR is determined based on Kabat numbering scheme. In some embodiments, the CDR is determined based on Chothia numbering scheme. In some embodiments, the CDR is determined based on a combination numbering scheme. In some embodiments, the CDR is determined based on IMGT numbering scheme.
- the disclosure also provides antibodies or antigen-binding fragments thereof that bind to TFR1 (human TFR1) .
- the antibodies or antigen-binding fragments thereof contain a heavy chain single variable region (VHH) comprising or consisting of an amino acid sequence that is at least 80%, 85%, 90%, or 95%identical to a selected VHH sequence.
- VHH heavy chain single variable region
- the selected VHH sequence is SEQ ID NO: 25.
- the selected VHH sequence is SEQ ID NO: 26.
- the selected VHH sequence is SEQ ID NO: 27.
- the selected VHH sequence is SEQ ID NO: 28.
- the sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in one or both of a first and a second amino acid or nucleic acid sequence for optimal alignment and non-homologous sequences can be disregarded for comparison purposes) .
- the amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared. When a position in the first sequence is occupied by the same amino acid residue or nucleotide as the corresponding position in the second sequence, then the molecules are identical at that position.
- the percent identity between the two sequences is a function of the number of identical positions shared by the sequences, taking into account the number of gaps, and the length of each gap, which need to be introduced for optimal alignment of the two sequences.
- the comparison of sequences and determination of percent identity between two sequences can be accomplished, e.g., using a Blossum 62 scoring matrix with a gap penalty of 12, a gap extend penalty of 4, and a frameshift gap penalty of 5.
- the disclosure also provides nucleic acid comprising a polynucleotide encoding a polypeptide comprising an immunoglobulin heavy chain single variable domain (VHH) .
- VHH comprises CDRs as shown in FIG. 1 and FIG. 2, or has sequences as shown in FIG. 3.
- the antibodies and antigen-binding fragments can also be antibody variants (including derivatives and conjugates) of antibodies or antibody fragments and multi-specific (e.g., bi-specific) antibodies or antibody fragments.
- Additional antibodies provided herein are polyclonal, monoclonal, multi-specific (multimeric, e.g., bi-specific) , human antibodies, chimeric antibodies (e.g., human-mouse chimera) , single-chain antibodies, intracellularly-made antibodies (i.e., intrabodies) , and antigen-binding fragments thereof.
- the antibodies or antigen-binding fragments thereof comprises an Fc domain that can be originated from various types (e.g., IgG, IgE, IgM, IgD, IgA, and IgY) , class (e.g., IgG1, IgG2, IgG3, IgG4, IgA1, and IgA2) , or subclass.
- the Fc domain is originated from an IgG antibody or antigen-binding fragment thereof.
- the Fc domain comprises one, two, three, four, or more heavy chain constant regions.
- the present disclosure also provides an antibody or antigen-binding fragment thereof that cross-competes with any antibody or antigen-binding fragment as described herein.
- the cross-competing assay is known in the art, and is described e.g., in Moore et al., "Antibody cross-competition analysis of the human immunodeficiency virus type 1 gp120 exterior envelope glycoprotein. " Journal of virology 70.3 (1996) : 1863-1872, which is incorporated herein reference in its entirety.
- the present disclosure also provides an antibody or antigen-binding fragment thereof that binds to the same epitope or region as any antibody or antigen-binding fragment as described herein.
- the epitope binning assay is known in the art, and is described e.g., in Estep et al. "High throughput solution-based measurement of antibody-antigen affinity and epitope binning. " MAbs. Vol. 5. No. 2. Taylor &Francis, 2013, which is incorporated herein reference in its entirety.
- the antibody or antigen-binding fragment thereof comprises a heavy chain single variable domain (VHH) CDR1 selected from SEQ ID NOs: 1, 4, 7, 10, 13, 16, 19, and 22.
- VHH heavy chain single variable domain
- the antibody or antigen-binding fragment thereof comprises a heavy chain single variable domain (VHH) CDR2 selected from SEQ ID NOs: 2, 5, 8, 11, 14, 17, 20, and 23.
- VHH heavy chain single variable domain
- the antibody or antigen-binding fragment thereof comprises a heavy chain single variable domain (VHH) CDR3 selected from SEQ ID NOs: 3, 6, 9, 12, 15, 18, 21, and 24.
- VHH heavy chain single variable domain
- TFR1 performs a critical role in cellular iron uptake through the interaction with iron-bound TF. Iron is required for multiple cellular processes and is essential for DNA synthesis and, thus, cellular proliferation. Due to its central role in cancer cell pathology, malignant cells often overexpress TFR1 and this increased expression can be associated with poor prognosis in different types of cancer. The elevated levels of TfR1 expression on malignant cells, together with its extracellular accessibility, ability to internalize, and central role in cancer cell pathology make this receptor an attractive target for antibody-mediated therapy.
- the antibodies or antigen-binding fragments thereof described herein cannot block the binding between TFR1 and TF. In some embodiments, the antibodies or antigen-binding fragments thereof described herein can block the binding between TFR1 and TF. In some embodiments, the antibodies or antigen-binding fragments thereof described herein can be conjugated to anti-cancer agents that are internalized by receptor-mediated endocytosis. In some embodiments, the antibodies or antigen-binding fragments thereof described herein can disrupt the function of the receptor. In some embodiments, the antibodies or antigen-binding fragments thereof described herein cannot induce Fc effector functions, therefore preventing or ameliorating their negative effects to normal cells.
- the disclosure provides antibodies or antigen-binding fragments thereof comprising a human Fc domain, which induce Fc-dependent effector functions by at least or about at least or about 1 fold, at least or about 2 folds, at least or about 3 folds, at least or about 4 folds, at least or about 5 folds, at least or about 6 folds, at least or about 7 folds, at least or about 8 folds, at least or about 9 folds, at least or about 10 folds, at least or about 20 folds, at least or about 30 folds, at least or about 40 folds, at least or about 50 folds, or at least or about 100 folds as compared when no antibodies or antigen-binding fragments thereof as described herein are present.
- the disclosure provides antibodies or antigen-binding fragments thereof comprising a human Fc domain, which induce host immune response by at least or about at least or about 1 fold, at least or about 2 folds, at least or about 3 folds, at least or about 4 folds, at least or about 5 folds, at least or about 6 folds, at least or about 7 folds, at least or about 8 folds, at least or about 9 folds, at least or about 10 folds, at least or about 20 folds, at least or about 30 folds, at least or about 40 folds, at least or about 50 folds, or at least or about 100 folds as compared when no antibodies or antigen-binding fragments thereof as described herein are present.
- the disclosure provides antibodies or antigen-binding fragments thereof that can internalize into human brain cells (e.g., cortical microvascular endothelial cells) that the endocytosis rate is at least 50%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%.
- human brain cells e.g., cortical microvascular endothelial cells
- the endocytosis rate of the antibodies or antigen-binding fragments thereof described herein is at least 1-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 50-fold, 100-fold, 500-fold, or 1000-fold as compared to that of an isotype control antibody.
- an antibody or antigen-binding fragment thereof comprising a single heavy chain. In some embodiments, provided herein is an antibody or antigen-binding fragment thereof comprising a pair of heavy chains. In some embodiments, the heavy chain pair is linked by disulfide bonds. In some embodiments, the heavy chain pair comprises knob-in-hole modifications. In some embodiments, the heavy chain comprises a human IgG Fc domain. In some embodiments, the antibody or antigen-binding fragment thereof comprises in each heavy chain at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 VHH domains. In some embodiments, the VHH domains in each heavy chain specifically bind to the same epitope. In some embodiments, the VHH domains in each heavy chain specifically bind to different epitopes. In some embodiments, the VHH domains in each heavy chain bind to at least 1, 2, 3, 4, or 5 different epitopes.
- the antibody or antigen-binding fragment thereof is a bi-specific antibody, or tri-specific antibody. In some embodiments, the antibody or antigen-binding fragment thereof can specifically bind to at least 4, 5, or 6 antigens.
- the antibody specifically binds to TFR1 with a dissociation rate (koff) of less than 0.1 s -1 , less than 0.01 s -1 , less than 0.001 s -1 , less than 0.0001 s -1 , or less than 0.00001 s -1 .
- the dissociation rate (koff) is greater than 0.01 s -1 , greater than 0.001 s -1 , greater than 0.0001 s -1 , greater than 0.00001 s -1 , or greater than 0.000001 s -1 .
- kinetic association rates (kon) is greater than 1 ⁇ 10 2 /Ms, greater than 1 ⁇ 10 3 /Ms, greater than 1 ⁇ 10 4 /Ms, greater than 1 ⁇ 10 5 /Ms, or greater than 1 ⁇ 10 6 /Ms. In some embodiments, kinetic association rates (kon) is less than 1 ⁇ 10 5 /Ms, less than 1 ⁇ 10 6 /Ms, or less than 1 ⁇ 10 7 /Ms.
- KD is less than 1 ⁇ 10 -6 M, less than 1 ⁇ 10 -7 M, less than 1 ⁇ 10 -8 M, less than 1 ⁇ 10 -9 M, or less than 1 ⁇ 10 -10 M. In some embodiments, the KD is less than 50 nM, 30 nM, 20 nM, 15 nM, 10 nM, 9 nM, 8 nM, 7 nM, 6 nM, 5 nM, 4 nM, 3 nM, 2 nM, or 1 nM.
- KD is greater than 1 x 10 -7 M, greater than 1 x 10 -8 M, greater than 1 x 10 -9 M, greater than 1 x 10 -10 M, greater than 1 x 10 -11 M, or greater than 1 x 10 -12 M.
- the antibody binds to human TFR1, monkey TFR1, mouse TFR1, or chimeric TFR1. In some embodiments, the antibody does not bind to human TFR1, monkey TFR1, mouse TFR1, or chimeric TFR1.
- thermal stabilities are determined.
- the antibodies or antigen-binding fragments as described herein can have a Tm (melting temperature) greater than 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, or 95 °C.
- Tm is less than 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, or 95 °C.
- the antibodies or antigen-binding fragments as described herein can have a Tagg (aggregation temperature, e.g., Tagg at 266 nm (Tagg266) or Tagg at 473 nm (Tagg473) ) great than 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, or 95 °C.
- Tagg aggregation temperature, e.g., Tagg at 266 nm (Tagg266) or Tagg at 473 nm (Tagg473)
- Tagg is less than 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, or 95 °C.
- the Fc region is human IgG1, human IgG2, human IgG3, or human IgG4.
- the antibodies or antigen binding fragments thereof have a functional Fc region.
- the antibodies or antigen binding fragments thereof comprise a human IgG1 Fc region.
- the human IgG1 Fc region comprises an amino acid sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identical to SEQ ID NO: 34.
- the antibodies or antigen binding fragments do not have an Fc region.
- the antibody (or antigen-binding fragment thereof) is a polypeptide comprising one or more VHH domains that are interconnected by linker peptides.
- the antibody comprises at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 VHH domain.
- the VHH domains specifically bind to the same epitope.
- the VHH domains bind to different epitopes.
- the VHH domains bind to at least 1, 2, 3, 4, or 5 different epitopes.
- the antibodies or antigen binding fragments thereof do not have a functional Fc region.
- the Fc region has LALA mutations (L234A and L235A mutations in EU numbering) , or LALA-PG mutations (L234A, L235A, P329G mutations in EU numbering) .
- the Fc region has a mutation at position 297 (e.g., N297A) according to EU numbering.
- the mutated human IgG1 Fc region comprises an amino acid sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identical to SEQ ID NO: 35.
- concentration of the antibody or antigen binding fragment thereof described herein in brain can be greater than 10%, 20%, 30%, 40%, 50%, 60%, 70%, or 80%of its concentration immediately (e.g., 0.5 hour) after administration.
- concentration of the antibody or antigen binding fragment thereof described herein in brain can be at least 1-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 20-fold, 30-fold, 40-fold, 50-fold, 60-fold, 70-fold, 80-fold, 90-fold, 100-fold, 200-fold, 300-fold, 400-fold, 500-fold, 1000-fold, 2000-fold, 5000-fold, or 10000-fold of the concentration of a control antibody (e.g., hIgG1 or JR141-N) , or the concentration in serum of the subject.
- a control antibody e.g., hIgG1 or JR141-N
- Variants of the antibodies or antigen-binding fragments described herein can be prepared by introducing appropriate nucleotide changes into the DNA encoding a human, humanized, or chimeric antibody, or antigen-binding fragment thereof described herein, or by peptide synthesis.
- Such variants include, for example, deletions, insertions, or substitutions of residues within the amino acids sequences that make-up the antigen-binding site of the antibody or an antigen-binding domain.
- some antibodies or antigen-binding fragments will have increased affinity for the target protein, e.g., TFR1.
- any combination of deletions, insertions, and/or combinations can be made to arrive at an antibody or antigen-binding fragment thereof that has increased binding affinity for the target.
- the amino acid changes introduced into the antibody or antigen-binding fragment can also alter or introduce new post-translational modifications into the antibody or antigen-binding fragment, such as changing (e.g., increasing or decreasing) the number of glycosylation sites, changing the type of glycosylation site (e.g., changing the amino acid sequence such that a different sugar is attached by enzymes present in a cell) , or introducing new glycosylation sites.
- the heavy-chain antibody or antigen-binding fragment thereof described herein is obtained by immunizing any of the genetically modified animals (e.g., mice with complete human heavy chain variable domain substitution in situ, combined with a modified constant region) described e.g., in PCT/CN2022/119188.
- the genetically modified animals e.g., mice with complete human heavy chain variable domain substitution in situ, combined with a modified constant region
- Humanized antibodies include antibodies having variable and constant regions derived from (or having the same amino acid sequence as those derived from) human germline immunoglobulin sequences of human immunoglobulin scaffold sequences. Humanized antibodies may include amino acid residues not encoded by human germline immunoglobulin sequences (e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo) . Accordingly, “humanized” antibodies are chimeric antibodies wherein sequences from a non-human species are substituted by the corresponding human sequences.
- amino acid sequence variants of the human, humanized, or chimeric anti-TFR1 antibody will contain an amino acid sequence having at least 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%percent identity with a sequence present in the VHH domain of the original antibody.
- Identity or homology with respect to an original sequence is usually the percentage of amino acid residues present within the candidate sequence that are identical with a sequence present within the human, humanized, or chimeric anti-TFR1 antibody or fragment, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity.
- a cysteine residue can be introduced into the Fc region, thereby allowing interchain disulfide bond formation in this region.
- the homodimeric antibody thus generated may have any increased half-life in vitro and/or in vivo.
- Homodimeric antibodies with increased half-life in vitro and/or in vivo can also be prepared using heterobifunctional cross-linkers as described, for example, in Wolff et al. Wolff et al. ( "Monoclonal antibody homodimers: enhanced antitumor activity in nude mice. " Cancer research 53.11 (1993) : 2560-2565) .
- an antibody can be engineered which has dual Fc regions.
- a covalent modification can be made to the anti-TFR1 antibody or antigen-binding fragment thereof.
- These covalent modifications can be made by chemical or enzymatic synthesis, or by enzymatic or chemical cleavage.
- Other types of covalent modifications of the antibody or antibody fragment are introduced into the molecule by reacting targeted amino acid residues of the antibody or fragment with an organic derivatization agent that is capable of reacting with selected side chains or the N-or C-terminal residues.
- antibody variants having a carbohydrate structure that lacks fucose attached (directly or indirectly) to an Fc region.
- the amount of fucose in such antibody composition may be from 1%to 80%, from 1%to 65%, from 5%to 65%or from 20%to 40%.
- the amount of fucose is determined by calculating the average amount of fucose within the sugar chain at Asn297, relative to the sum of all glycostructures attached to Asn 297 (e.g. complex, hybrid and high mannose structures) as measured by MALDI-TOF mass spectrometry, as described in WO 2008/077546, for example.
- Asn297 refers to the asparagine residue located at about position 297 in the Fc region (Eu numbering of Fc region residues; or position 314 in Kabat numbering) ; however, Asn297 may also be located about ⁇ 3 amino acids upstream or downstream of position 297, i.e., between positions 294 and 300, due to minor sequence variations in antibodies. Such fucosylation variants may have improved ADCC function.
- the Fc region of the antibody can be further engineered to replace the Asparagine at position 297 with Alanine (N297A) .
- the present disclosure also provides recombinant vectors (e.g., an expression vectors) that include an isolated polynucleotide disclosed herein (e.g., a polynucleotide that encodes a polypeptide disclosed herein) , host cells into which are introduced the recombinant vectors (i.e., such that the host cells contain the polynucleotide and/or a vector comprising the polynucleotide) , and the production of recombinant antibody polypeptides or fragments thereof by recombinant techniques.
- recombinant vectors e.g., an expression vectors
- an isolated polynucleotide disclosed herein e.g., a polynucleotide that encodes a polypeptide disclosed herein
- host cells into which are introduced the recombinant vectors (i.e., such that the host cells contain the polynucleotide and/or a vector comprising the polynucleo
- a “vector” is any construct capable of delivering one or more polynucleotide (s) of interest to a host cell when the vector is introduced to the host cell.
- An “expression vector” is capable of delivering and expressing the one or more polynucleotide (s) of interest as an encoded polypeptide in a host cell into which the expression vector has been introduced.
- the polynucleotide of interest is positioned for expression in the vector by being operably linked with regulatory elements such as a promoter, enhancer, and/or a poly-Atail, either within the vector or in the genome of the host cell at or near or flanking the integration site of the polynucleotide of interest such that the polynucleotide of interest will be translated in the host cell introduced with the expression vector.
- regulatory elements such as a promoter, enhancer, and/or a poly-Atail
- a vector can be introduced into the host cell by methods known in the art, e.g., electroporation, chemical transfection (e.g., DEAE-dextran) , transformation, transfection, and infection and/or transduction (e.g., with recombinant virus) .
- vectors include viral vectors (which can be used to generate recombinant virus) , naked DNA or RNA, plasmids, cosmids, phage vectors, and DNA or RNA expression vectors associated with cationic condensing agents.
- a polynucleotide disclosed herein e.g., a polynucleotide that encodes a polypeptide disclosed herein
- a viral expression system e.g., vaccinia or other pox virus, retrovirus, or adenovirus
- vaccinia or other pox virus, retrovirus, or adenovirus may involve the use of a non-pathogenic (defective) , replication competent virus, or may use a replication defective virus.
- viral propagation generally will occur only in complementing virus packaging cells. Suitable systems are disclosed, for example, in Fisher-Hoch et al., 1989, Proc. Natl. Acad. Sci. USA 86: 317-321; Flexner et al., 1989, Ann. N. Y.
- the DNA insert comprising an antibody-encoding or polypeptide-encoding polynucleotide disclosed herein can be operatively linked to an appropriate promoter (e.g., a heterologous promoter) , such as the phage lambda PL promoter, the E. coli lac, trp and tac promoters, the SV40 early and late promoters and promoters of retroviral LTRs, to name a few. Other suitable promoters are known to the skilled artisan.
- the promoter is a cytomegalovirus (CMV) promoter.
- CMV cytomegalovirus
- the expression constructs can further contain sites for transcription initiation, termination and, in the transcribed region, a ribosome binding site for translation.
- the coding portion of the mature transcripts expressed by the constructs may include a translation initiating at the beginning and a termination codon (UAA, UGA, or UAG) appropriately positioned at the end of the polypeptide to be translated.
- the expression vectors can include at least one selectable marker.
- markers include dihydrofolate reductase or neomycin resistance for eukaryotic cell culture and tetracycline or ampicillin resistance genes for culturing in E. coli and other bacteria.
- Representative examples of appropriate hosts include, but are not limited to, bacterial cells, such as E. coli, Streptomyces, and Salmonella typhimurium cells; fungal cells, such as yeast cells; insect cells such as Drosophila S2 and Spodoptera Sf9 cells; animal cells such as CHO, COS, Bowes melanoma, and HK 293 cells; and plant cells. Appropriate culture mediums and conditions for the host cells described herein are known in the art.
- Non-limiting vectors for use in bacteria include pQE70, pQE60 and pQE-9, available from Qiagen; pBS vectors, Phagescript vectors, Bluescript vectors, pNH8A, pNH16a, pNH18A, pNH46A, available from Stratagene; and ptrc99a, pKK223-3, pKK233-3, pDR540, pRIT5 available from Pharmacia.
- Non-limiting eukaryotic vectors include pWLNEO, pSV2CAT, pOG44, pXT1 and pSG available from Stratagene; and pSVK3, pBPV, pMSG and pSVL available from Pharmacia. Other suitable vectors will be readily apparent to the skilled artisan.
- Non-limiting bacterial promoters suitable for use include the E. coli lacI and lacZ promoters, the T3 and T7 promoters, the gpt promoter, the lambda PR and PL promoters and the trp promoter.
- Suitable eukaryotic promoters include the CMV immediate early promoter, the HSV thymidine kinase promoter, the early and late SV40 promoters, the promoters of retroviral LTRs, such as those of the Rous sarcoma virus (RSV) , and metallothionein promoters, such as the mouse metallothionein-I promoter.
- yeast Saccharomyces cerevisiae a number of vectors containing constitutive or inducible promoters such as alpha factor, alcohol oxidase, and PGH can be used.
- Introduction of the construct into the host cell can be affected by calcium phosphate transfection, DEAE-dextran mediated transfection, cationic lipid-mediated transfection, electroporation, transduction, infection or other methods.
- Such methods are described in many standard laboratory manuals, such as Davis et al., Basic Methods in Molecular Biology (1986) , which is incorporated herein by reference in its entirety.
- Enhancers are cis-acting elements of DNA, usually about from 10 to 300 bp that act to increase transcriptional activity of a promoter in a given host cell-type.
- enhancers include the SV40 enhancer, which is located on the late side of the replication origin at base pairs 100 to 270, the cytomegalovirus early promoter enhancer, the polyoma enhancer on the late side of the replication origin, and adenovirus enhancers.
- secretion signals may be incorporated into the expressed polypeptide.
- the signals may be endogenous to the polypeptide or they may be heterologous signals.
- the polypeptide (e.g., antibody) can be expressed in a modified form, such as a fusion protein (e.g., a GST-fusion) or with a histidine-tag, and may include not only secretion signals, but also additional heterologous functional regions. For instance, a region of additional amino acids, particularly charged amino acids, may be added to the N-terminus of the polypeptide to improve stability and persistence in the host cell, during purification, or during subsequent handling and storage. Also, peptide moieties can be added to the polypeptide to facilitate purification. Such regions can be removed prior to final preparation of the polypeptide. The addition of peptide moieties to polypeptides to engender secretion or excretion, to improve stability and to facilitate purification, among others, are familiar and routine techniques in the art.
- the disclosure also provides a nucleic acid sequence that is at least 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%identical to any nucleotide sequence as described herein, and an amino acid sequence that is at least 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%identical to any amino acid sequence as described herein.
- the disclosure relates to nucleotide sequences encoding any peptides that are described herein, or any amino acid sequences that are encoded by any nucleotide sequences as described herein.
- the nucleic acid sequence is less than 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 150, 200, 250, 300, 350, 400, 500, or 600 nucleotides.
- the amino acid sequence is less than 5, 6, 7, 8, 9, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, or 200 amino acid residues.
- the amino acid sequence (i) comprises an amino acid sequence; or (ii) consists of an amino acid sequence, wherein the amino acid sequence is any one of the sequences as described herein.
- the nucleic acid sequence (i) comprises a nucleic acid sequence; or (ii) consists of a nucleic acid sequence, wherein the nucleic acid sequence is any one of the sequences as described herein.
- the antibody or antigen-binding fragment thereof is expressed in yeast, insect cells, or mammalian cells (e.g., CHO cells) .
- the disclosure provides methods for treating a brain disease (e.g., brain cancer, dementia, or Alzheimer’s disease) in a subject, methods of identifying a subject having a brain disease (e.g., brain cancer, dementia, or Alzheimer’s disease) , methods of reducing the risk of developing a brain disease, or methods of reducing the risk of developing additional symptoms in a subject.
- the treatment can halt, slow, retard, or inhibit progression of a brain disease (e.g., brain cancer, dementia, or Alzheimer’s disease) .
- the treatment can result in the reduction of in the number, severity, and/or duration of one or more symptoms of a brain disease (e.g., brain cancer, dementia, or Alzheimer’s disease) in a subject.
- the disclosure features methods that include administering a therapeutically effective amount of an antibody or antigen-binding fragment thereof disclosed herein to a subject in need thereof (e.g., a subject having, or identified or diagnosed as having, a brain disease) .
- the disclosure features methods that carry a therapeutic agent to cross the blood brain barrier.
- the antibodies or antigen-binding fragments thereof as described herein are linked to the therapeutic agent.
- the therapeutic agent is an antibody, an antigen binding fragment thereof, a small molecule, or an antibody-drug conjugate.
- compositions and methods disclosed herein can be used for treatment of patients at risk for a brain disease (e.g., brain cancer, dementia, or Alzheimer’s disease) .
- a brain disease e.g., brain cancer, dementia, or Alzheimer’s disease
- Patients with a brain disease can be identified with various methods known in the art.
- the brain disease is a brain cancer.
- the disclosure is related to methods of decreasing the rate of tumor growth, including contacting a tumor cell with an effective amount of a composition including the antibody or antigen-binding fragment thereof, or the antibody-drug conjugate described herein. In one aspect, the disclosure is related to methods of killing a tumor cell, including contacting a tumor cell with an effective amount of a composition including the antibody or antigen-binding fragment thereof, or the antibody-drug conjugate described herein.
- an “effective amount” is meant an amount or dosage sufficient to effect beneficial or desired results including halting, slowing, retarding, or inhibiting progression of a disease, e.g., a cancer.
- An effective amount will vary depending upon, e.g., an age and a body weight of a subject to which the antibody, antigen binding fragment, antibody-encoding polynucleotide, vector comprising the polynucleotide, and/or compositions thereof is to be administered, a severity of symptoms and a route of administration, and thus administration can be determined on an individual basis.
- an effective amount can be administered in one or more administrations.
- an effective amount of an antibody or an antigen binding fragment thereof is an amount sufficient to ameliorate, stop, stabilize, reverse, inhibit, slow and/or delay progression of a disease in a patient.
- an effective amount of an antibody or antigen binding fragment may vary, depending on, inter alia, patient history as well as other factors such as the type (and/or dosage) of antibody used.
- Effective amounts and schedules for administering the antibodies, antibody-encoding polynucleotides, and/or compositions disclosed herein may be determined empirically, and making such determinations is within the skill in the art. Those skilled in the art will understand that the dosage that must be administered will vary depending on, for example, the mammal that will receive the antibodies, antibody-encoding polynucleotides, and/or compositions disclosed herein, the route of administration, the particular type of antibodies, antibody-encoding polynucleotides, antigen binding fragments, and/or compositions disclosed herein used and other drugs being administered to the mammal.
- a typical daily dosage of an effective amount of an antibody is 0.01 mg/kg to 100 mg/kg.
- the dosage can be less than 100 mg/kg, 10 mg/kg, 9 mg/kg, 8 mg/kg, 7 mg/kg, 6 mg/kg, 5 mg/kg, 4 mg/kg, 3 mg/kg, 2 mg/kg, 1 mg/kg, 0.5 mg/kg, or 0.1 mg/kg.
- the dosage can be greater than 10 mg/kg, 9 mg/kg, 8 mg/kg, 7 mg/kg, 6 mg/kg, 5 mg/kg, 4 mg/kg, 3 mg/kg, 2 mg/kg, 1 mg/kg, 0.5 mg/kg, 0.1 mg/kg, 0.05 mg/kg, or 0.01 mg/kg.
- the dosage is about 10 mg/kg, 9 mg/kg, 8 mg/kg, 7 mg/kg, 6 mg/kg, 5 mg/kg, 4 mg/kg, 3 mg/kg, 2 mg/kg, 1 mg/kg, 0.9 mg/kg, 0.8 mg/kg, 0.7 mg/kg, 0.6 mg/kg, 0.5 mg/kg, 0.4 mg/kg, 0.3 mg/kg, 0.2 mg/kg, or 0.1 mg/kg.
- the at least one antibody, antigen-binding fragment thereof, or pharmaceutical composition e.g., any of the antibodies, antigen-binding fragments, or pharmaceutical compositions described herein
- at least one additional therapeutic agent can be administered to the subject at least once a week (e.g., once a week, twice a week, three times a week, four times a week, once a day, twice a day, or three times a day) .
- at least two different antibodies and/or antigen-binding fragments are administered in the same composition (e.g., a liquid composition) .
- At least one antibody or antigen-binding fragment and at least one additional therapeutic agent are administered in the same composition (e.g., a liquid composition) .
- the at least one antibody or antigen-binding fragment and the at least one additional therapeutic agent are administered in two different compositions (e.g., a liquid composition containing at least one antibody or antigen-binding fragment and a solid oral composition containing at least one additional therapeutic agent) .
- the at least one additional therapeutic agent is administered as a pill, tablet, or capsule.
- the at least one additional therapeutic agent is administered in a sustained-release oral formulation.
- the one or more additional therapeutic agents can be administered to the subject prior to, or after administering the at least one antibody, antigen-binding antibody fragment, or pharmaceutical composition (e.g., any of the antibodies, antigen-binding antibody fragments, or pharmaceutical compositions described herein) .
- the one or more additional therapeutic agents and the at least one antibody, antigen-binding antibody fragment, or pharmaceutical composition are administered to the subject such that there is an overlap in the bioactive period of the one or more additional therapeutic agents and the at least one antibody or antigen-binding fragment (e.g., any of the antibodies or antigen-binding fragments described herein) in the subject.
- the subject can be administered the at least one antibody, antigen-binding antibody fragment, or pharmaceutical composition (e.g., any of the antibodies, antigen- binding antibody fragments, or pharmaceutical compositions described herein) over an extended period of time (e.g., over a period of at least 1 week, 2 weeks, 3 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 12 months, 1 year, 2 years, 3 years, 4 years, or 5 years) .
- a skilled medical professional may determine the length of the treatment period using any of the methods described herein for diagnosing or following the effectiveness of treatment (e.g., the observation of at least one symptom of the disease) .
- a skilled medical professional can also change the identity and number (e.g., increase or decrease) of antibodies or antigen-binding antibody fragments (and/or one or more additional therapeutic agents) administered to the subject and can also adjust (e.g., increase or decrease) the dosage or frequency of administration of at least one antibody or antigen-binding antibody fragment (and/or one or more additional therapeutic agents) to the subject based on an assessment of the effectiveness of the treatment (e.g., using any of the methods described herein and known in the art) .
- one or more additional therapeutic agents can be administered to the subject.
- the additional therapeutic agent can comprise one or more inhibitors selected from the group consisting of an inhibitor of B-Raf, an EGFR inhibitor, an inhibitor of a MEK, an inhibitor of ERK, an inhibitor of K-Ras, an inhibitor of c-Met, an inhibitor of anaplastic lymphoma kinase (ALK) , an inhibitor of a phosphatidylinositol 3-kinase (PI3K) , an inhibitor of an Akt, an inhibitor of mTOR, a dual PI3K/mTOR inhibitor, an inhibitor of Bruton's tyrosine kinase (BTK) , and an inhibitor of Isocitrate dehydrogenase 1 (IDH1) and/or Isocitrate dehydrogenase 2 (IDH2) .
- Isocitrate dehydrogenase 1 IDH1
- Isocitrate dehydrogenase 2 IDH2
- the additional therapeutic agent can comprise one or more inhibitors selected from the group consisting of an inhibitor of HER3, an inhibitor of LSD1, an inhibitor of MDM2, an inhibitor of BCL2, an inhibitor of CHK1, an inhibitor of activated hedgehog signaling pathway, and an agent that selectively degrades the estrogen receptor.
- the additional therapeutic agent can comprise one or more therapeutic agents selected from the group consisting of Trabectedin, nab-paclitaxel, Trebananib, Pazopanib, Cediranib, Palbociclib, everolimus, fluoropyrimidine, IFL, regorafenib, Reolysin, Alimta, Zykadia, Sutent, temsirolimus, axitinib, everolimus, sorafenib, Votrient, Pazopanib, IMA-901, AGS-003, cabozantinib, Vinflunine, an Hsp90 inhibitor, Ad-GM-CSF, Temazolomide, IL-2, IFNa, vinblastine, Thalomid, dacarbazine, cyclophosphamide, lenalidomide, azacytidine, lenalidomide, bortezomid, amrubicine, carfilzomib, prala
- therapeutic agents
- the additional therapeutic agent can comprise one or more therapeutic agents selected from the group consisting of an adjuvant, a TLR agonist, tumor necrosis factor (TNF) alpha, IL-1, HMGB1, an IL-10 antagonist, an IL-4 antagonist, an IL-13 antagonist, an IL-17 antagonist, an HVEM antagonist, an ICOS agonist, a treatment targeting CX3CL1, a treatment targeting CXCL9, a treatment targeting CXCL10, a treatment targeting CCL5, an LFA-1 agonist, an ICAM1 agonist, and a Selectin agonist.
- TNF tumor necrosis factor
- carboplatin, nab-paclitaxel, paclitaxel, cisplatin, pemetrexed, gemcitabine, FOLFOX, or FOLFIRI are administered to the subject.
- the additional therapeutic agent is an anti-PD-1 antibody, an anti-PD-L1 antibody, an anti-LAG-3 antibody, an anti-TIGIT antibody, an anti-4-1BB antibody, an anti-CTLA-4 antibody, an anti-CD40 antibody, an anti-OX40 antibody, or an anti-GITR antibody.
- compositions that contain at least one (e.g., one, two, three, or four) of the antibodies or antigen-binding fragments described herein. Two or more (e.g., two, three, or four) of any of the antibodies or antigen-binding fragments described herein can be present in a pharmaceutical composition in any combination.
- the pharmaceutical compositions may be formulated in any manner known in the art.
- compositions are formulated to be compatible with their intended route of administration (e.g., intravenous, intraarterial, intramuscular, intradermal, subcutaneous, or intraperitoneal) .
- the compositions can include a sterile diluent (e.g., sterile water or saline) , a fixed oil, polyethylene glycol, glycerine, propylene glycol or other synthetic solvents, antibacterial or antifungal agents, such as benzyl alcohol or methyl parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like, antioxidants, such as ascorbic acid or sodium bisulfite, chelating agents, such as ethylenediaminetetraacetic acid, buffers, such as acetates, citrates, or phosphates, and isotonic agents, such as sugars (e.g., dextrose) , polyalcohols (e.g., mannitol or
- Liposomal suspensions can also be used as pharmaceutically acceptable carriers (see, e.g., U.S. Patent No. 4,522,811) .
- Preparations of the compositions can be formulated and enclosed in ampules, disposable syringes, or multiple dose vials. Where required (as in, for example, injectable formulations) , proper fluidity can be maintained by, for example, the use of a coating, such as lecithin, or a surfactant.
- Absorption of the antibody or antigen-binding fragment thereof can be prolonged by including an agent that delays absorption (e.g., aluminum monostearate and gelatin) .
- controlled release can be achieved by implants and microencapsulated delivery systems, which can include biodegradable, biocompatible polymers (e.g., ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid; Alza Corporation and Nova Pharmaceutical, Inc. ) .
- biodegradable, biocompatible polymers e.g., ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid; Alza Corporation and Nova Pharmaceutical, Inc.
- compositions containing one or more of any of the antibodies or antigen-binding fragments described herein can be formulated for parenteral (e.g., intravenous, intraarterial, intramuscular, intradermal, subcutaneous, or intraperitoneal) administration in dosage unit form (i.e., physically discrete units containing a predetermined quantity of active compound for ease of administration and uniformity of dosage) .
- parenteral e.g., intravenous, intraarterial, intramuscular, intradermal, subcutaneous, or intraperitoneal
- dosage unit form i.e., physically discrete units containing a predetermined quantity of active compound for ease of administration and uniformity of dosage
- Toxicity and therapeutic efficacy of compositions can be determined by standard pharmaceutical procedures in cell cultures or experimental animals (e.g., monkeys) .
- Agents that exhibit high therapeutic indices are preferred. Where an agent exhibits an undesirable side effect, care should be taken to minimize potential damage (i.e., reduce unwanted side effects) .
- Toxicity and therapeutic efficacy can be determined by other standard pharmaceutical procedures.
- a therapeutically effective amount of the one or more (e.g., one, two, three, or four) antibodies or antigen-binding fragments thereof (e.g., any of the antibodies or antibody fragments described herein) will be an amount that treats the disease in a subject in a subject, or a subject identified as being at risk of developing the disease, decreases the severity, frequency, and/or duration of one or more symptoms of a disease in a subject (e.g., a human) .
- any of the antibodies or antigen-binding fragments described herein can be determined by a health care professional or veterinary professional using methods known in the art, as well as by the observation of one or more symptoms of disease in a subject (e.g., a human) . Certain factors may influence the dosage and timing required to effectively treat a subject (e.g., the severity of the disease or disorder, previous treatments, the general health and/or age of the subject, and the presence of other diseases) .
- Exemplary doses include milligram or microgram amounts of any of the antibodies or antigen-binding fragments described herein per kilogram of the subject’s weight (e.g., about 1 ⁇ g/kg to about 500 mg/kg; about 100 ⁇ g/kg to about 500 mg/kg; about 100 ⁇ g/kg to about 50 mg/kg; about 10 ⁇ g/kg to about 5 mg/kg; about 10 ⁇ g/kg to about 0.5 mg/kg; or about 1 ⁇ g/kg to about 50 ⁇ g/kg) . While these doses cover a broad range, one of ordinary skill in the art will understand that therapeutic agents, including antibodies and antigen-binding fragments thereof, vary in their potency, and effective amounts can be determined by methods known in the art.
- relatively low doses are administered at first, and the attending health care professional or veterinary professional (in the case of therapeutic application) or a researcher (when still working at the development stage) can subsequently and gradually increase the dose until an appropriate response is obtained.
- the specific dose level for any particular subject will depend upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, gender, and diet of the subject, the time of administration, the route of administration, the rate of excretion, and the half-life of the antibody or antibody fragment in vivo.
- compositions can be included in a container, pack, or dispenser together with instructions for administration.
- disclosure also provides methods of manufacturing the antibodies or antigen binding fragments thereof for various uses as described herein.
- RenNano TM mice Biocytogen, complete human heavy chain variable domain substitution in situ, combined with a modified constant region. The mice are described e.g., in PCT/CN2022/119188, which is incorporated herein by reference in its entirety) were immunized with His-tagged human TFR1 (transferrin receptor 1) protein (hTFR1-His, ACROBiosystems, Cat#: CD1-H5243) to obtain anti-TFR1 antibodies.
- retro-orbital blood was collected as a negative control.
- Freund’s complete adjuvant (CFA) was used for the first immunization
- Freund’s incomplete adjuvant (IFA) was used for the second and third immunizations. A total of three immunizations (bi-weekly) were performed.
- One week after the third immunization retro-orbital blood was collected, and the antibody titer of serum was detected by FACS.
- TFR1 protein was injected by intraperitoneal injection, and the CHO-Scells expressing human TFR1 antigen was injected through the tail vein.
- Antigen-specific immune cells were isolated from the immunized mice to further obtain anti-TFR1 antibodies or to obtain the heavy chain variable region sequences of the anti-TFR1 antibodies.
- single cell technology e.g., using Optofluidic System, Berkeley Lights Inc.
- Reverse transcription and PCR sequencing were used to obtain antibody variable region sequences.
- the obtained variable region sequences were used for antibody expression to verify the binding activity to TFR1 using FACS. Because the lack of the CH1 domain, the heavy chain variable region (VH) of the obtained antibodies is also referred to as a heavy chain single variable domain (VHH) .
- VHH sequences were respectively connected to a human IgG1 constant region (e.g., the hinge region, CH2 domain and CH3 domain) .
- exemplary antibodies obtained by this method included: 23B8, 24A1, 24C9 and 24G5.
- the heavy chain CDR1-3 sequences are shown in FIG. 1 and FIG. 2.
- the VHH region sequences of 23B8, 24A1, 24C9 and 24G5 are shown in FIG. 3.
- the constant region of the antibodies can be further engineered to replace the Asparagine at position 297 with Alanine (N297A) .
- N297A mutation is introduced into the constant region of 24G5
- the resulting antibody is named as 24G5-N.
- one-armed antibodies 23B8-mono, 24A1-mono, 24C9-mono and 24G5-mono were constructed, these antibodies have an anti-TFR1 arm comprising a VHH region, and a heavy chain fragment comprising CH2 and CH3 domains of IgG1 with N297A mutation.
- CHO-S-hTFR1 cells or CHO-S-fasTFR1 cells were transferred to a 96-well plate at a density of 10 5 cells/well respectively.
- Serially diluted sample anti-TFR1 antibodies were added to the 96-well plate, and incubated at 4°C for 30 minutes.
- PBS was used as a negative control (NC) .
- the cells were incubated with the secondary antibody anti-hIgG-Fc-Alex Flour TM 647 (Jackson ImmunoResearch Laboratories, Cat#: 109-606-170) at 4°C in the dark for 15 minutes before flow cytometry analysis.
- CHO-S-hTFR1 cells or CHO-S-fasTFR1 cells were obtained by transfecting CHO-Scells with vectors expressing human TFR1 (hTFR1, SEQ ID NO: 29) or Macaca fascicularis (crab-eating macaque) TFR1 amino acid sequence (fasTFR1, SEQ ID NO: 30) , respectively.
- the test results are shown in the table below.
- JR141 a humanized IgG1 antibody targeting human TFR1 conjugated to human iduronate-2-sulfatase, was first approved in March 2021 in Japan for the intravenous treatment of mucopolysaccharidosis type II.
- the VH and VL sequences of JR141 are set forth in SEQ ID NO: 31 and SEQ ID NO: 32, respectively.
- positive control JR141-N
- the VH and VL of JR141 were connected to a human IgG1 constant region with N297A mutation.
- the binding affinity of the anti-TFR1 antibodies to His-tagged TFR1 protein of human (hTFR1-His, ACROBiosystems, Cat#: CD1-H5243) or monkey (fasTFR1-His, ACROBiosystems, Cat#: TFR-C524a) were verified using surface plasmon resonance (SPR) on Biacore TM (Biacore, Inc., Piscataway N. J. ) 8K biosensor equipped with pre-immobilized Protein A sensor chips.
- SPR surface plasmon resonance
- Purified anti-TFR1 antibodies was captured on the Protein A chip (Series S Sensor Chip Protein A) for the detection.
- Purified anti-TFR1 antibodies (1 ⁇ g/mL) was loaded at 10 ⁇ L/min to bind to hTFR1-His and fasTFR1-His (200 nM) .
- the flow rate was 30 ⁇ L/min.
- the binding and dissociation time were set to 180 seconds and 600 seconds, respectively.
- the chip was regenerated after the last injection of each titration with a glycine solution (pH 2.0) at 30 ⁇ L/min for 30 seconds.
- HBS-EP+ buffer 10 mM 4- (2-hydroxyethyl) -1-piperazineethanesulfonic acid (HEPES) , 150 mM NaCl, 3 mM ethylenediaminetetraacetic acid (EDTA) and 0.05%P20, pH7.4) diluted from HBS-EP+ buffer (10 ⁇ ) was used as the running buffer throughout the experiment.
- HBS-EP+ buffer 10 mM 4- (2-hydroxyethyl) -1-piperazineethanesulfonic acid (HEPES) , 150 mM NaCl, 3 mM ethylenediaminetetraacetic acid (EDTA) and 0.05%P20, pH7.4
- HIS1K Anti-Penta-HIS
- Analyte 1 was injected at a flow rate of 30 ⁇ L/min to bind the ligand.
- Another antibody was injected under the same conditions to determine whether the binding of different antibodies interfered with each other. The binding time was 300 seconds for each antibody.
- the binding value of each antibody was obtained using Data Analysis HT 12.0. To quantify the interference of one antibody binding to another, a binding ratio was calculated to compare each pair of antibodies. The binding ratio is defined as the binding value of the second antibody (analyte 2) , divided by the binding value of the first antibody (analyte 1) . The binding ratio of each antibody pair is summarized in the matrix table below. Specifically, the binding ratio was between 0.0 to 0.5, if analyte 1 exhibited a blocking effect to analyte 2; the binding ratio was between 0.5-1.1, if analyte 1 did not exhibit a blocking effect to analyte 2. In general, antibody pairs that interfere with each other have the same or overlapping epitopes.
- the epitope binding assay results indicate that 24A1 and 24G5 can recognize the same epitope, and 23B8, 24C9 and JR141-N can recognize different epitopes.
- Anti-TFR1 antibodies together with the pHAb-Goat anti-human IgG secondary antibody were added to human cortical microvascular endothelial cells (hCMEC/D3 cells) , and incubated for 3 hours. After incubation, the cells were centrifuged and washed with FACS buffer. Mean fluorescence intensity (MFI) was measured using a flow cytometer. Endocytosis rates of antibodies were calculated.
- Human IgG1 protein (CrownBio, Cat#: C0001) was used as an isotype control (ISO) . The results are shown in the following table, indicating that all four antibodies exhibited good endocytosis activity in human cortical microvascular endothelial cells.
- the Agilent 1290 chromatograph system (connected with XBridge TM Protein BEH SEC column ( Waters Corporation) ) was used.
- the antibody samples were diluted to 1 mg/mL with purified water.
- the following parameters were used: mobile phase: 25 mM phosphate buffer (PB) (pH 6.8) + 0.3 M NaCl; flow rate: 1.8 mL/min; column temperature: 25 °C; detection wavelength: 280 nm; injection volume: 10 ⁇ L; sample tray temperature: 6 °C; and running time: 7 minutes.
- PB phosphate buffer
- flow rate 1.8 mL/min
- column temperature 25 °C
- detection wavelength 280 nm
- injection volume 10 ⁇ L
- sample tray temperature 6 °C
- running time 7 minutes.
- mobile phase A 0.9 M ammonium sulfate, 0.1 M PB, 10%acetonitrile pH 6.5
- mobile phase B 0.1 M PB, 10%acetonitrile pH 6.5
- flow rate 0.8 mL/min
- gradient 0 min 100%A, 2 min 100%A, 32 min 100%B, 34 min 100%B, 35 min 100%A, and 45 min 100%A
- column temperature 30 °C
- detection wavelength 280 nm
- injection volume 10 ⁇ g
- sample tray temperature about 6 °C
- running time 45 minutes.
- the Maurice cIEF Method Development Kit (Protein Simple, Cat#: PS-MDK01-C) was used for sample preparation. Specifically, 40 ⁇ g protein sample was mixed with the following reagents in the kit: 1 ⁇ L Maurice cIEF pI Marker-4.05, 1 ⁇ L Maurice cIEF pI Marker-9.99, 35 ⁇ L 1%Methyl Cellulose Solution, 2 ⁇ L Maurice cIEF 500 mM Arginine, 4 ⁇ L Ampholytes (Pharmalyte pH ranges 3-10) , and water (added to make a final volume of 100 ⁇ L) .
- Maurice cIEF Cartridges PS-MC02-C were used to generate imaging capillary isoelectric focusing spectra. The sample was focused for a total of 10 minutes. The analysis software installed on the instrument was used to analyze the absorbance of the 280 nm-focused protein.
- the antibodies were diluted to 1 mg/mL using PBS buffer, and the following tests were performed: (1) detecting the specificity of the antibodies using the Cross-Interaction Chromatography (CIC) method (indicated as the retention time (CIC, min) ) ; (2) detecting the colloidal stabilities of the antibodies by the stand-up monolayer chromatography (SMAC) method (indicated as the retention time (SMAC, %/min) ) .
- CIC Cross-Interaction Chromatography
- SMAC stand-up monolayer chromatography
- a CIC column was prepared by coupling human polyclonal IgGs (Sigma, Cat #: I4506) onto a HiTrap NHS-activated resin (GE Healthcare, Cat #: 17-0716-01) followed by passivation with ethanolamine according to published procedures.
- the column was then connected to Agilent 1260 chromatograph system and run at 0.1 mL/min using 1 ⁇ PBS as the mobile phase until a flat baseline was reached. 10 ⁇ g of antibodies at 1 mg/mL in PBS were then injected. Peak retention times on the column were monitored at 280 nm; running time: 50 minutes.
- Zenix column (4.6 mm ⁇ 30 cm, Sepax, Cat #: 213300-4630) was connected to the column compartment, and place the appropriate line in the mobile phase. Equilibrate column for 60 min at 0.350 mL/min flow rate with mobile phase buffer. Load antibodies into the injection sequence.
- Mobile Phase A 150 mM Sodium Phosphate pH 7.0; flow rate: 0.35 mL/min; running time: 25 min; column temperature: 30°C; detection wavelength: 280 nm, 220 nm.
- a humanized TFR1 mouse model (hTFR1 mice) was engineered to express a chimeric TFR1 protein (SEQ ID NO: 33) in which the extracellular region of mouse TFR1 protein was replaced with the corresponding human TFR1 extracellular region.
- SEQ ID NO: 33 a chimeric TFR1 protein in which the extracellular region of mouse TFR1 protein was replaced with the corresponding human TFR1 extracellular region.
- mice The concentrations of the anti-TFR1 antibodies were determined in hTFR1 mice. Specifically, the mice were placed into different groups (8 mice per group) , and administered with an approximately equal molar dosage of JR141-N (G2) , 23B8-N (G3) , 24A1-N (G4) , 24G5-N (G5) , or 24C9-N (G6) , by intravenous (i. v. ) injection.
- the control group (G1) mice were administered with human IgG1 (hIgG1) . Details of the administration scheme are shown in the table below.
- mice Blood and brain samples were collected 0.5, 6, 24, and 72 hours after the administration. Two mice were sampled at each time point, and the mice were anesthetized after retro-orbital blood was collected. To avoid interference from the residual blood in the brain, the mice were perfused by saline for 10 minutes at room temperature. Specifically, saline was perfused via systemic circulation from the left ventricle to the right ventricle. Brain samples were excised and divided into two hemibrains by the sagittal plane. The left hemibrain was subjected to quantification of the injected antibody, while the right hemibrain was fixed by formalin and embedded with paraffin for serial sections.
- FIGS. 4A-4D show the antibody concentration in total brain protein (FIG. 4A) , the ratio of antibody concentration in brain total protein to serum antibody concentration (FIG.
- hTFR1 mice were placed into five groups (3 mice per group) and administered with 18.4 mg/kg JR141-N (G2) , 10 mg/kg 23B8-N (G3) , 10 mg/kg 24A1-N (G4) , or 10 mg/kg 24G5-N (G5) by intravenous injection (1 administration in total) .
- the control group (G1) mice were administered with hIgG1 (G1) .
- brain samples were collected to determine the concentrations of the anti-TFR1 antibodies.
- FIGS. 5A-5B show the antibody concentration test results in brain parenchyma and brain total protein, respectively.
- hTFR1 mice were placed into seven groups (6 mice per group) and administered with JR141-N (G2-G4) or 24G5-N (G5-G7) ) by intravenous (i. v. ) injection.
- the control group (G1) mice were administered with hIgG1. Details of the administration scheme are shown in the table below.
- HBS-EP+ buffer 10 mM 4- (2-hydroxyethyl) -1-piperazineethanesulfonic acid (HEPES) , 150 mM NaCl, 3 mM EDTA and 0.05%Surfactant P20, pH 7.4
- HBS-EP+ buffer 10 mM 4- (2-hydroxyethyl) -1-piperazineethanesulfonic acid (HEPES) , 150 mM NaCl, 3 mM EDTA and 0.05%Surfactant P20, pH 7.4
- Each purified antibody (23B8-N, 24A1-N, 24C9-N, 24G5-N, 23B8-mono, 24A1-mono, 24C9-mono and 24G5-mono) was coupled with Dxd (Deruxtecan) through GGFG linker.
- ADC is added directly after the antibody name.
- 24G5-N is coupled to GGFG-Dxd, it is named as 24G5-ADC.
- 24G5-mono is coupled to GGFG-Dxd, it is named as 24G5-mono-ADC.
- HIC-HPLC Reversed Phase High Performance Liquid Chromatography
- hTFR1 mice were placed into different groups (6 mice per group) and administered with 10 mg/kg bivalent anti-TFR1 ADCs (24G5-ADC, 23B8-ADC, 24A1-ADC, 24C9-ADC) or the same molar dose 8.34 mg/kg one-armed monovalent anti-TFR1 ADCs (23B8-mono-ADC, 24A1-mono-ADC, 24C9-mono-ADC, or 24G5-mono-ADC) by intravenous (i. v. ) injection (1 administration in total) . 0.5 hour, 18 hours and 72 hours after the administration, blood and brain samples were collected using the methods described in Example 7.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Engineering & Computer Science (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Medicinal Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Animal Behavior & Ethology (AREA)
- Veterinary Medicine (AREA)
- Pharmacology & Pharmacy (AREA)
- Immunology (AREA)
- Epidemiology (AREA)
- Organic Chemistry (AREA)
- Cell Biology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- General Chemical & Material Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Biophysics (AREA)
- Hematology (AREA)
- Oncology (AREA)
- Molecular Biology (AREA)
- Genetics & Genomics (AREA)
- Biochemistry (AREA)
- Peptides Or Proteins (AREA)
Abstract
The present disclosure provides an anti-TFR1 (transferrin receptor 1) antibodies, antigen-binding fragments, and the uses thereof.
Description
CLAIM OF PRIORITY
This application claims the benefit of PCT Application No. PCT/CN2022/136246, filed on December 2, 2022. The entire contents of the foregoing are incorporated herein by reference.
This disclosure relates to anti-TFR1 (transferrin receptor 1) antibodies, antigen-binding fragments, and the uses thereof.
Therapeutic antibodies are one of the fastest growing classes of therapeutic compounds, rapidly outpacing the growth of small-molecule drugs. For example, monoclonal antibodies have revolutionized cancer therapy. However, delivery to tumor cells in vivo is hampered by the large size of conventional antibodies. The minimal target recognition module of a conventional antibody is composed of two non-covalently associated variable domains (VH and VL) . The inherent hydrophobic interaction of VH and VL domains limits the stability and solubility of engineered antibodies, often causing aggregation and/or mispairing of V-domains.
The discovery of heavy-chain antibodies has given rise to unprecedented opportunities in impacting cancer therapy. These unique forms of camelid-derived antibodies lack the entire light chain and the CH1 domain and are only composed of a single variable domain termed VHH. Recombinant VHHs are small (15-20 kDa) and strictly monomeric; they bind their target with nM affinity as well as with being stable in a broad pH and temperature ranges. Molecular manipulation is also easier with VHH; this facilitates the production of multivalent formats of monoclonal antibodies compared with conventional recombinant antibodies and their fragments, which is problematic due to aggregation and reduced affinity. Moreover, VHH often binds to epitopes that are less immunogenic for conventional antibodies.
Usually, the therapeutic antibodies are human or humanized antibodies. The human or humanized antibodies can be generated by humanization of a rodent antibody (e.g., a mouse antibody) or by using phage libraries. However, these animals or phage libraries usually cannot produce heavy chain antibodies. Instead, heavy chain antibodies are often obtained from camelid heavy chain antibodies. These camelid heavy chain antibodies need to be humanized. The humanization process may adversely affect the binding affinity and introduce immunogenic epitopes to the antibodies. Iterative and time-consuming experiments are often required to improve the properties of these antibodies. And in some cases, these antibodies can also be immunogenic in patients, leading to attenuation of their efficacy over time. Therefore, there is a need to develop more forms of antibodies to treat or prevent human diseases.
The present disclosure relates to an antibody or antigen-binding fragment thereof that binds to transferrin receptor 1 (TFR1) , comprising: a heavy chain single variable domain (VHH) comprising complementarity determining regions (CDRs) 1, 2, and 3, in some embodiments, the VHH CDR1 region comprises an amino acid sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identical to a selected VHH CDR1 amino acid sequence, the VHH CDR2 region comprises an amino acid sequence that is at least 80%, 85%, 90%, 95%,
96%, 97%, 98%, 99%, or 100%identical to a selected VHH CDR2 amino acid sequence, and the VHH CDR3 region comprises an amino acid sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identical to a selected VHH CDR3 amino acid sequence; in some embodiments, the selected VHH CDRs 1, 2, and 3 amino acid sequences are one of the following:
(1) the selected VHH CDRs 1, 2, 3 amino acid sequences are set forth in SEQ ID NOs: 1, 2, and 3, respectively;
(2) the selected VHH CDRs 1, 2, 3 amino acid sequences are set forth in SEQ ID NOs: 4, 5, and 6, respectively;
(3) the selected VHH CDRs 1, 2, 3 amino acid sequences are set forth in SEQ ID NOs: 7, 8, and 9, respectively;
(4) the selected VHH CDRs 1, 2, 3 amino acid sequences are set forth in SEQ ID NOs: 10, 11, and 12, respectively;
(5) the selected VHH CDRs 1, 2, 3 amino acid sequences are set forth in SEQ ID NOs: 13, 14, and 15, respectively;
(6) the selected VHH CDRs 1, 2, 3 amino acid sequences are set forth in SEQ ID NOs: 16, 17, and 18, respectively;
(7) the selected VHH CDRs 1, 2, 3 amino acid sequences are set forth in SEQ ID NOs: 19, 20, and 21, respectively; and
(8) the selected VHH CDRs 1, 2, 3 amino acid sequences are set forth in SEQ ID NOs: 22, 23, and 24, respectively.
In some embodiments, the VHH comprises CDRs 1, 2, 3 with the amino acid sequences set forth in SEQ ID NOs: 1, 2, and 3, respectively. In some embodiments, the VHH comprises CDRs 1, 2, 3 with the amino acid sequences set forth in SEQ ID NOs: 4, 5, and 6, respectively. In some embodiments, the VHH comprises CDRs 1, 2, 3 with the amino acid sequences set forth in SEQ ID NOs: 7, 8, and 9, respectively. In some embodiments, the VHH comprises CDRs 1, 2, 3 with the amino acid sequences set forth in SEQ ID NOs: 10, 11, and 12, respectively.
In one aspect, the disclosure is related to an antibody or antigen-binding fragment thereof that binds to TFR1 comprising a heavy chain single variable region (VHH) comprising an amino acid sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identical to a selected VHH sequence, in some embodiments, the selected VHH sequence is selected from the group consisting of SEQ ID NOs: 25, 26, 27, and 28. In some embodiments, the VHH comprises the sequence of SEQ ID NO: 25. In some embodiments, the VHH comprises the sequence of SEQ ID NO: 26. In some embodiments, the VHH comprises the sequence of SEQ ID NO: 27. In some embodiments, the VHH comprises the sequence of SEQ ID NO: 28. In some embodiments, the antibody or antigen-binding fragment specifically binds to a human TFR1, a monkey TFR1, a mouse TFR1, or a chimeric TFR1. In some embodiments, the antibody or antigen-binding fragment is a human or humanized antibody or antigen-binding fragment thereof. In some embodiments, the antibody or antigen-binding fragment is a multi-specific antibody (e.g., a bispecific antibody) .
In one aspect, the disclosure is related to an antibody or antigen-binding fragment thereof comprising the VHH CDRs 1, 2, 3, of the antibody or antigen-binding fragment thereof as described herein.
In some embodiments, the antibody or antigen-binding fragment comprises a human IgG Fc (e.g., a human IgG1 Fc) . In some embodiments, the human IgG Fc comprises a non-asparagine residue (e.g., alanine) at position 297 according to EU numbering. In some
embodiments, the antibody or antigen-binding fragment comprises two or more heavy chain single variable domains.
In one aspect, the disclosure is related to a nucleic acid comprising a polynucleotide encoding the antibody or antigen-binding fragment thereof as described herein. In some embodiments, the nucleic acid is cDNA.
In one aspect, the disclosure is related to a vector comprising one or more of the nucleic acids as described herein.
In one aspect, the disclosure is related to a cell comprising the vector as described herein. In some embodiments, the cell is a CHO cell. In one aspect, the disclosure is related to a cell comprising one or more of the nucleic acids described herein.
In one aspect, the disclosure is related to a method of producing an antibody or an antigen-binding fragment thereof, the method comprising (a) culturing the cell as described herein under conditions sufficient for the cell to produce the antibody or the antigen-binding fragment thereof; and (b) collecting the antibody or the antigen-binding fragment thereof produced by the cell.
In one aspect, the disclosure is related to an antibody-drug conjugate comprising the antibody or antigen-binding fragment thereof as described herein covalently bound to a therapeutic agent. In some embodiments, the therapeutic agent is a cytotoxic or cytostatic agent.
In one aspect, the disclosure is related to a method of treating a subject having a brain disease (e.g., a brain cancer) , the method comprising administering a therapeutically effective amount of a composition comprising the antibody or antigen-binding fragment thereof, or the antibody-drug conjugate as described herein, to the subject. In some embodiments, the antibody or antigen-binding fragment thereof, or the antibody-drug conjugate can pass across the blood-brain barrier (BBB) of the subject.
In one aspect, the disclosure is related to a method of treating a subject having a cancer, the method comprising administering a therapeutically effective amount of a composition comprising the antibody or antigen-binding fragment thereof, or the antibody-drug conjugate as described herein, to the subject. In some embodiments, the cancer is brain cancer, lung cancer, gastric cancer, colorectal cancer, liver cancer, ovarian cancer, prostate cancer, leukemia, or breast cancer. In one aspect, the disclosure is related to a method of identifying a subject as having a brain disease (e.g., a brain cancer) , the method comprising detecting a sample collected from the subject as having the brain disease by the antibody or antigen-binding fragment thereof as described herein, thereby identifying the subject as having the brain disease. In some embodiments, the sample is a brain parenchyma sample from the subject. In some embodiments, the subject described herein is a human subject.
In one aspect, the disclosure is related to a method of delivering an agent to cross blood brain barrier, the method comprising administering the agent covalently linked to the antibody or antigen-binding fragment thereof as described herein to the subject. In some embodiments, the agent is an antibody or an antibody drug conjugate. In some embodiments, the agent is anti-amyloid antibody.
In one aspect, the disclosure is related to a pharmaceutical composition comprising the antibody or antigen-binding fragment thereof as described herein, and a pharmaceutically acceptable carrier. In one aspect, the disclosure is related to a pharmaceutical composition comprising the antibody drug conjugate as described herein, and a pharmaceutically acceptable carrier.
In one aspect, the disclosure is related to an antibody or antigen-binding fragment thereof that cross-competes with the antibody or antigen-binding fragment thereof as described herein.
In one aspect, the disclosure provides a method of making an antibody that specifically binds to an antigen. The method involves exposing the animal as described herein to the antigen; obtaining the sequence of (e.g. by sequencing) nucleic acids encoding human heavy chain immunoglobulin variable regions in a cell that expresses a chimeric heavy chain antibody that specifically binds to the antigen; and operably linking in a cell the nucleic acid encoding the human heavy chain immunoglobulin variable region with a nucleic acid encoding a human heavy chain immunoglobulin constant region.
The disclosure also relates to an offspring of the non-human mammal. In some embodiments, the non-human mammal is a rodent. In some embodiments, the non-human mammal is a mouse.
The disclosure also provides to a cell including the targeting vector as described herein. The disclosure also relates to a cell (e.g., a stem cell, an embryonic stem cell, an immune cell, a B cell, a T cell, or a hybridoma) or a cell line, or a primary cell culture thereof derived from the non-human mammal or an offspring thereof. The disclosure further relates to the tissue, organ or a culture thereof derived from the non-human mammal or an offspring thereof.
The disclosure further relates to the use of the non-human mammal or an offspring thereof, the animal model generated through the method as described herein in the development of a product related to an immunization process, the manufacture of a human antibody, or the model system for research in pharmacology, immunology, microbiology and medicine.
Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Methods and materials are described herein for use in the present invention; other, suitable methods and materials known in the art can also be used. The materials, methods, and examples are illustrative only and not intended to be limiting. All publications, patent applications, patents, sequences, database entries, and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including definitions, will control.
Other features and advantages of the invention will be apparent from the following detailed description and figures, and from the claims.
FIG. 1 lists CDR sequences of heavy chain variable region of anti-TFR1 antibodies according to Kabat numbering.
FIG. 2 lists CDR sequences of heavy chain variable region of anti-TFR1 antibodies according to IMGT numbering.
FIG. 3 lists amino acids sequences discussed in the disclosure.
FIG. 4A shows the antibody concentration in total brain protein of hTFR1 mice within 72 hours of intravenous (i. v. ) administration of hIgG1 (G1) , JR141-N (G2) , 23B8-N (G3) , 24A1-N (G4) , 24G5-N (G5) , or 24C9-N (G6) .
FIG. 4B shows the ratio of antibody concentration in brain total protein to serum antibody concentration of hTFR1 mice within 72 hours of intravenous (i. v. ) administration of hIgG1 (G1) , JR141-N (G2) , 23B8-N (G3) , 24A1-N (G4) , 24G5-N (G5) , or 24C9-N (G6) .
FIG. 4C shows the antibody concentration in brain parenchyma of hTFR1 mice within 72 hours of intravenous (i. v. ) administration of hIgG1 (G1) , JR141-N (G2) , 23B8-N (G3) , 24A1-N (G4) , 24G5-N (G5) , or 24C9-N (G6) .
FIG. 4D shows the ratio of antibody concentration in brain parenchyma to serum antibody concentration of hTFR1 mice within 72 hours of intravenous (i. v. ) administration of hIgG1 (G1) , JR141-N (G2) , 23B8-N (G3) , 24A1-N (G4) , 24G5-N (G5) , or 24C9-N (G6) .
FIG. 5A shows the antibody concentration test results in brain parenchyma after 24 hours of intravenous (i. v. ) administration of hIgG1 (G1) , JR141-N (G2) , 23B8-N (G3) , 24A1-N (G4) , or 24G5-N (G5) .
FIG. 5B shows the antibody concentration test results in brain total protein (Whole Brain) after 24 hours of intravenous (i. v. ) administration of hIgG1 (G1) , JR141-N (G2) , 23B8-N (G3) , 24A1-N (G4) , or 24G5-N (G5) .
FIG. 6 shows the antibody concentration results after 6 hours or 24 hours of intravenous (i.v. ) administration of JR141-N (G2-G4) or 24G5-N (G5-G7) ) . hIgG1 was used as a negative control.
FIG. 7 shows the ADC concentration test results in brain parenchyma after 72 hours of intravenous (i. v. ) administration of 24G5-ADC or 24G5-mono-ADC.
A heavy-chain antibody (or heavy chain-only antibody) is an antibody which has only heavy chains (generally two heavy chains) and lacks the two light chains usually found in antibodies. Naturally occurring heavy-chain antibodies have been discovered in cartilaginous fishes (e.g., shark) and camelids (e.g., llama) . For example, in cartilaginous fishes, the immunoglobulin new antigen receptor (IgNAR) is a heavy-chain antibody. IgNAR shows significant structural differences to other antibodies. It has five constant domains (CH) per chain instead of the usual three, several disulfide bonds in unusual positions, and the complementarity-determining region 3 (CDR3) forms an extended loop covering the site which binds to a light chain in other antibodies. These differences, in combination with the phylogenetic age of the cartilaginous fishes, have led to the hypothesis that IgNAR could be more closely related to a primordial antigen-binding protein than the mammalian immunoglobulins.
The only mammals with heavy-chain (IgG-like) antibodies are camelids such as dromedaries, camels, llamas and alpacas. Like all mammals, camelids (e.g., llamas) can produce conventional antibodies made of two heavy chains and two light chains bound together with disulfide bonds in a Y shape (e.g., IgG1) . However, they also produce two unique subclasses of IgG: IgG2 and IgG3, also known as heavy chain IgG. These antibodies are made of only two heavy chains, which lack the CH1 region but still bear an antigen-binding domain (e.g., VHH) at their N-terminus. Conventional Ig require the association of variable regions from both heavy and light chains to allow a high diversity of antigen-antibody interactions. Although isolated heavy and light chains still show this capacity, they exhibit very low affinity when compared to paired heavy and light chains. The unique feature of heavy chain IgG is the capacity of their monomeric antigen binding regions to bind antigens with specificity, affinity and especially diversity that are comparable to conventional antibodies without the need of pairing with another region. This feature is mainly due to a couple of major variations within the amino acid sequence of the variable region of the two heavy chains, which induce deep conformational changes when compared to conventional Ig. Major substitutions in the variable regions prevent the light chains
from binding to the heavy chains, but also prevent unbound heavy chains from being recycled by the Immunoglobulin Binding Protein.
The single variable domain of these heavy-chain antibodies (designated VHH, sdAb, or nanobody) is the smallest antigen-binding domain generated by adaptive immune systems. The Complementarity Determining Region 3 (CDR3) of the variable region of these antibodies has often been found to be twice as long as the conventional ones. This results in an increased interaction surface with the antigen as well as an increased diversity of antigen-antibody interactions, which compensates the absence of the light chains. With a long complementarity-determining region 3 (CDR3) , VHHs can extend into crevices on proteins that are not accessible to conventional antibodies, including functionally interesting sites such as the active site of an enzyme or the receptor-binding canyon on a virus surface. Moreover, an additional cysteine residue allows the structure to be more stable, thus increasing the strength of the interaction.
VHHs offer numerous other advantages compared to conventional antibodies carrying variable domains (VH and VL) of conventional antibodies, including higher stability, solubility, expression yields, and refolding capacity, as well as better in vivo tissue penetration. Moreover, in contrast to the VH domains of conventional antibodies, VHH do not display an intrinsic tendency to bind to light chains. This facilitates the induction of heavy chain antibodies in the presence of a functional light chain loci. Further, since VHH do not bind to VL domains, it is much easier to reformat VHHs into bispecific antibody constructs than constructs containing conventional VH-VL pairs or single domains based on VH domains.
A notable difference between the camelid VHH and the human VH domain is the length and orientation of the CDR3 loop. The CDR3 corresponds to the unique region of the antibody molecule that is encoded by a DNA element newly generated during B-cell development. Genetic recombination results in the fusion of a D-element with flanking V-and J-elements. During recombination further genetic diversity is generated by addition and/or deletion of nucleotides at the junctions. Thereby, the CDR3 loop provides the major contribution to antibody diversity and specificity. A limited number of variable region genes (IGHV, IGHD, and IGHJ) in some early transgenic heavy chain antibody animals, results in some antigens not being recognized by these animals, despite potent antigen response by wildtype animals (Janssens, Rick, et al. "Generation of heavy-chain-only antibodies in mice. " Proceedings of the National Academy of Sciences 103.41 (2006) : 15130-15135) . The present disclosure provides fully humanized heavy chain antibodies generated by genetically modified animals that have complete human heavy chain antibody repertoires..
As used herein, the term “antibody” refers to any antigen-binding molecule that contains at least one (e.g., one, two, three, four, five, or six) complementary determining region (CDR) (e.g., any of the three CDRs from an immunoglobulin light chain or any of the three CDRs from an immunoglobulin heavy chain) and is capable of specifically binding to an epitope. Non-limiting examples of antibodies include: monoclonal antibodies, polyclonal antibodies, multi-specific antibodies (e.g., bi-specific antibodies) , single-chain antibodies, heavy chain antibodies, chimeric antibodies, human antibodies, and humanized antibodies. In some embodiments, an antibody can contain an Fc region of a human antibody. The term antibody also includes derivatives, e.g., bi-specific antibodies, single-chain antibodies, diabodies, linear antibodies, and multi-specific antibodies formed from antibody fragments.
As used herein, the term “antigen-binding fragment” refers to a portion of a full-length antibody, wherein the portion is capable of specifically binding to an antigen. In some embodiments, the antigen-binding fragment contains at least one variable domain (e.g., a
variable domain of a heavy chain or a variable domain of light chain) . Non-limiting examples of antibody fragments include, e.g., Fab, Fab’, F (ab’) 2, and Fv fragments.
As used herein, the term “human antibody” refers to an antibody that is encoded by a nucleic acid (e.g., rearranged human immunoglobulin heavy or light chain locus) present in a human. In some embodiments, a human antibody is collected from a human or produced in a human cell culture (e.g., human hybridoma cells) . In some embodiments, a human antibody is produced in a non-human cell (e.g., a mouse or hamster cell line) . In some embodiments, a human antibody is produced in a bacterial or yeast cell. In some embodiments, a human antibody is produced in a transgenic non-human animal (e.g., a mouse) containing an unrearranged or rearranged human immunoglobulin locus (e.g., heavy or light chain human immunoglobulin locus) .
As used herein, the term “chimeric antibody” refers to an antibody that contains a sequence present in at least two different antibodies (e.g., antibodies from two different mammalian species such as a human and a mouse antibody) . A non-limiting example of a chimeric antibody is an antibody containing the variable domain sequences (e.g., all or part of a light chain and/or heavy chain variable domain sequence) of a human antibody and the constant domains of a non-human antibody. Additional examples of chimeric antibodies are described herein and are known in the art.
As used herein, the term “humanized antibody” refers to a non-human antibody which contains sequence derived from a non-human (e.g., mouse) immunoglobulin and contains sequences derived from a human immunoglobulin.
As used herein, the term “single-chain antibody” refers to a single polypeptide that contains at least two immunoglobulin variable domains (e.g., a variable domain of a mammalian immunoglobulin heavy chain or light chain) that is capable of specifically binding to an antigen.
As used herein, the term “heavy-chain antibody” refers to an antibody molecule which is composed only of heavy chains (generally two) and does not have any light chains.
As used herein, the term “VHH” refers to the variable domain derived from a heavy-chain antibody. The VHH can specifically recognize an antigen without the need to be paired with a VL. In some embodiments, the VHH (also know as sdAb or nanobody) described herein is derived from any of the humanized heavy-chain antibody described herein. In some embodiments, the VHH, sdAb, or nanobody described herein is derived from the heavy chain antibody produced by any of the genetically modified non-human animal described herein.
As used herein, the terms “subject” and “patient” are used interchangeably throughout the specification and describe an animal, human or non-human. Veterinary and non-veterinary applications are contemplated by the present disclosure. Human patients can be adult humans or juvenile humans (e.g., humans below the age of 18 years old) . In addition to humans, patients include but are not limited to mice, rats, hamsters, guinea-pigs, rabbits, ferrets, cats, dogs, and primates. Included are, for example, non-human primates (e.g., monkey, chimpanzee, gorilla, and the like) , rodents (e.g., rats, mice, gerbils, hamsters, ferrets, rabbits) , lagomorphs, swine (e.g., pig, miniature pig) , equine, canine, feline, bovine, and other domestic, farm, and zoo animals.
As used herein, when referring to an antibody, the phrases “specifically binding” and “specifically binds” mean that the antibody interacts with its target molecule preferably to other molecules, because the interaction is dependent upon the presence of a particular structure (i.e., the antigenic determinant or epitope) on the target molecule; in other words, the reagent is recognizing and binding to molecules that include a specific structure rather than to all molecules
in general. An antibody that specifically binds to the target molecule may be referred to as a target-specific antibody.
As used herein, the terms “polypeptide, ” “peptide, ” and “protein” are used interchangeably to refer to polymers of amino acids of any length of at least two amino acids.
As used herein, the terms “polynucleotide, ” “nucleic acid molecule, ” and “nucleic acid sequence” are used interchangeably herein to refer to polymers of nucleotides of any length of at least two nucleotides, and include, without limitation, DNA, RNA, DNA/RNA hybrids, and modifications thereof.
Antibodies and Antigen Binding Fragments
The present disclosure provides antibodies and antigen-binding fragments thereof (e.g., heavy chain antibodies, humanized heavy chain antibodies, or multi-specific antibodies) that are produced by the methods described herein.
In general, conventional antibodies are made up of two classes of polypeptide chains, light chains and heavy chains. A non-limiting antibody of the present disclosure can be an intact, four-immunoglobulin-chain antibody comprising two heavy chains and two light chains. The heavy chain of the antibody can be of any isotype including IgM, IgG, IgE, IgA, or IgD or subclasses including IgG1, IgG2, IgG2a, IgG2b, IgG3, IgG4, IgE1, IgE2, etc. The light chain can be a kappa light chain or a lambda light chain. An antibody can comprise two identical copies of a light chain and two identical copies of a heavy chain. The heavy chains, which each contain one variable domain (or variable region, VH) and multiple constant domains (or constant regions) , bind to one another via disulfide bonding within their constant domains to form the “stem” of the antibody. The light chains, which each contain one variable domain (or variable region, VL) and one constant domain (or constant region) , each bind to one heavy chain via disulfide binding. The variable region of each light chain is aligned with the variable region of the heavy chain to which it is bound. The variable regions of both the light chains and heavy chains contain three hypervariable regions sandwiched between more conserved framework regions (FR) .
These hypervariable regions, known as the complementary determining regions (CDRs) , form loops that comprise the principle antigen binding surface of the antibody. The four framework regions largely adopt a beta-sheet conformation and the CDRs form loops connecting, and in some cases forming part of, the beta-sheet structure. The CDRs in each chain are held in close proximity by the framework regions and, with the CDRs from the other chain, contribute to the formation of the antigen-binding region.
Methods for identifying the CDR regions of an antibody by analyzing the amino acid sequence of the antibody are well known, and a number of definitions of the CDRs are commonly used. The Kabat definition is based on sequence variability, and the Chothia definition is based on the location of the structural loop regions. These methods and definitions are described in, e.g., Martin, "Protein sequence and structure analysis of antibody variable domains, " Antibody engineering, Springer Berlin Heidelberg, 2001.422-439; Abhinandan, et al. "Analysis and improvements to Kabat and structurally correct numbering of antibody variable domains, " Molecular immunology 45.14 (2008) : 3832-3839; Wu, T. T. and Kabat, E. A. (1970) J. Exp. Med. 132: 211-250; Martin et al., Methods Enzymol. 203: 121-53 (1991) ; Morea et al., Biophys Chem. 68 (1-3) : 9-16 (Oct. 1997) ; Morea et al., J Mol Biol. 275 (2) : 269-94 (Jan . 1998) ; Chothia et al., Nature 342 (6252) : 877-83 (Dec. 1989) ; Ponomarenko and Bourne, BMC Structural Biology 7: 64 (2007) ; each of which is incorporated herein by reference in its entirety.
The CDRs are important for recognizing an epitope of an antigen. As used herein, an “epitope” is the smallest portion of a target molecule capable of being specifically bound by the antigen binding domain of an antibody. The minimal size of an epitope may be about three, four, five, six, or seven amino acids, but these amino acids need not be in a consecutive linear sequence of the antigen’s primary structure, as the epitope may depend on an antigen’s three-dimensional configuration based on the antigen’s secondary and tertiary structure.
In some embodiments, the antibody is an intact immunoglobulin molecule (e.g., IgG1, IgG2a, IgG2b, IgG2c, IgG3, IgG4, IgM, IgD, IgE, IgA) . The IgG subclasses (IgG1, IgG2, IgG3, and IgG4) are highly conserved, differ in their constant region, particularly in their hinges and upper CH2 domains. The sequences and differences of the IgG subclasses are known in the art, and are described, e.g., in Vidarsson, et al, "IgG subclasses and allotypes: from structure to effector functions. " Frontiers in immunology 5 (2014) ; Irani, et al. "Molecular properties of human IgG subclasses and their implications for designing therapeutic monoclonal antibodies against infectious diseases. " Molecular immunology 67.2 (2015) : 171-182; Shakib, Farouk, ed. The human IgG subclasses: molecular analysis of structure, function and regulation. Elsevier, 2016; each of which is incorporated herein by reference in its entirety. The heavy chain constant regions in the heavy chain antibodies can be derived from any immunoglobulin molecules described herein (e.g., IgG1, IgG2a, IgG2b, IgG2c, IgG3, IgG4, IgM, IgD, IgE, IgA) .
The antibody can also be an immunoglobulin molecule that is derived from any species (e.g., human, rodent, mouse, rat, camelid) . Antibodies disclosed herein also include, but are not limited to, polyclonal, monoclonal, monospecific, multi-specific antibodies, and chimeric antibodies that include an immunoglobulin binding domain fused to another polypeptide. The term “antigen binding domain” or “antigen binding fragment” is a portion of an antibody that retains specific binding activity of the intact antibody, i.e., any portion of an antibody that is capable of specific binding to an epitope on the intact antibody’s target molecule. It includes, e.g., Fab, Fab', F (ab') 2, and variants of these fragments. Thus, in some embodiments, an antibody or an antigen binding fragment thereof can be, e.g., a scFv, a Fv, a Fd, a dAb, a bispecific antibody, a bispecific scFv, a diabody, a linear antibody, a single-chain antibody molecule, a multi-specific antibody formed from antibody fragments, and any polypeptide that includes a binding domain which is, or is homologous to, an antibody binding domain. Non-limiting examples of antigen binding domains include, e.g., the heavy chain and/or light chain CDRs of an intact antibody, the heavy and/or light chain variable regions of an intact antibody, full length heavy or light chains of an intact antibody, or an individual CDR from either the heavy chain or the light chain of an intact antibody.
In some embodiments, the antigen binding fragment can form a part of a chimeric antigen receptor (CAR) . In some embodiments, the chimeric antigen receptor are fusions of VHH as described herein, fused to CD3-zeta transmembrane-and endodomain.
The antibodies and antigen-binding fragments thereof (e.g., humanized antibodies or chimeric antibodies) that are produced by the methods described herein have various advantages. In some embodiments, no further optimization is required to obtain desired properties (e.g., binding affinities, thermal stabilities, and/or limited aggregation) .
In some implementations, the antibody (or antigen-binding fragments thereof) specifically binds to a target with a dissociation rate (koff) of less than 0.1 s-1, less than 0.01 s-1, less than 0.001 s-1, less than 0.0001 s-1, or less than 0.00001 s-1. In some embodiments, the dissociation rate (koff) is greater than 0.01 s-1, greater than 0.001 s-1, greater than 0.0001 s-1, greater than 0.00001 s-1, or greater than 0.000001 s-1.
In some embodiments, kinetic association rates (kon) are greater than 1 x 102/Ms, greater than 1 x 103/Ms, greater than 1 x 104/Ms, greater than 1 x 105/Ms, or greater than 1 x 106/Ms. In some embodiments, kinetic association rates (kon) are less than 1 x 105/Ms, less than 1 x 106/Ms, or less than 1 x 107/Ms.
Affinities can be deduced from the quotient of the kinetic rate constants (KD=koff/kon) . In some embodiments, KD is less than 1 x 10-6 M, less than 1 x 10-7 M, less than 1 x 10-8 M, less than 1 x 10-9 M, or less than 1 x 10-10 M. In some embodiments, the KD is less than 50 nM, 40 nM, 30 nM, 20 nM, 15 nM, 10 nM, 9 nM, 8 nM, 7 nM, 6 nM, 5 nM, 4 nM, 3 nM, 2 nM, or 1 nM. In some embodiments, KD is greater than 1 x 10-7 M, greater than 1 x 10-8 M, greater than 1 x 10-
9 M, greater than 1 x 10-10 M, greater than 1 x 10-11 M, or greater than 1 x 10-12 M. In some embodiments, the antibody binds to a target with KD less than or equal to about 0.9 nM, 0.8 nM, 0.7 nM, 0.6 nM, 0.5 nM, 0.4 nM, 0.3 nM, 0.2 nM, or 0.1 nM.
In some embodiments, thermal stabilities are determined. The antibodies or antigen binding fragments as described herein can have a Tm greater than 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, or 95 ℃.
In various embodiments, substitutions are performed to a parental heavy chain antibody sequence to make a variant heavy chain antibody. In general, a heavy chain antibody variant of a parental heavy chain antibody has an antigen binding affinity that is at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%or at least 100% (e.g., at least 150%, at least 200%, at least 500%, at least 1000%, or up to at least 10,000%) of the binding affinity of the parental heavy chain antibody to a particular antigen. In some embodiments, a variant heavy chain antibody will comprise a single substitution as compared to a parental heavy chain antibody. However, in other embodiments, several amino acids, e.g., up to about 5 or 10 or more, are substituted as compared to the parental heavy chain antibody sequence that are derived from other human heavy chain sequences that share identity at a given position. In various embodiments, the resultant variant heavy chain antibody is tested to confirm that the desired binding affinity and/or specificity has not been significantly decreased by the replacement residues. In some embodiments, an improved variant heavy chain antibody is produced by the substitution of amino acids from a different human heavy chain sequence. In various embodiments, the VHH is at least 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%identical to a parental VHH.
The VHH described herein can be used to make multi-specific (e.g., bispecific antibodies) . In one aspect, the present disclosure provides a multi-specific antibody comprising: a first antigen binding portion and a second antigen binding portion. In some embodiments, the first antigen binding portion comprises a heavy chain variable domain (VH) and a light chain variable domain (VL) , wherein the VH and VL together form an antigen-binding site that specifically binds a first epitope. In some embodiments, the first antigen binding portion comprises a VHH that specifically binds a first epitope. In some embodiments, the second antigen binding portion comprises a VHH that specifically binds a second epitope. In some embodiments, the first epitope and the second epitope are from the same antigen. In some embodiments, the first epitope and the second epitope are from different antigens.
In some embodiments, the first antigen binding portion is a full-length antibody consisting of two heavy chains and two light chains. In some embodiments, the first antigen binding portion is an antibody fragment comprising a heavy chain comprising the VH and a light chain comprising the VL. In some embodiments, the second antigen binding portion comprises a
single polypeptide chain. In some embodiments, the C terminus of the second antigen binding portion is fused to the N-terminus of at least one heavy chain of the first antigen binding portion. In some embodiments, the C terminus of the second antigen binding portion is fused to the N-terminus of at least one light chain of the first antigen binding portion. In some embodiments, the N terminus of the second antigen binding portion is fused to the C-terminus of at least one heavy chain of the first antigen binding portion. In some embodiments, the N terminus of the second antigen binding portion is fused to the C-terminus of at least one light chain of the first antigen binding portion. In some embodiments, the second antigen binding portion is a Fab-like domain comprising a first polypeptide chain comprising a first VHH fused to a CH1 domain, and a second polypeptide chain comprising a second VHH fused to a CL domain.
In some embodiments, the antibody or antigen-binding fragment thereof is a tri-specific antibody. In some embodiments, the tri-specific antibody is a tri-specific VHH-Fc. In some embodiments, the tri-specific antibody comprises the same VHHs. In some embodiments, the tri-specific antibody comprises different VHHs. In some embodiments, the VHHs bind to the same epitope. In some embodiments, the VHHs bind to different epitopes.
In some embodiments, the antibody or antigen-binding fragment thereof has four or more than four VHHs. In some embodiments, in order to increase developability, at least four VHHs are combined without the addition of IgG Fc domain to construct tetra-specific VHHs. These molecules would have the added advantage of increased affinity and avidity towards the antigen compared to bi-and tri-specific VHH-Fcs, despite lacking the Fc effector functions.
In some embodiments, these the antibody or antigen-binding fragment thereof (e.g., comprising 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more than 10) has a functional Fc.
In some embodiments, the heavy-chain antibody produced by the genetically modified non-human animal described herein has a VHH domain that includes CDR1, CDR2, and CDR3. In some embodiments, the CDR3 length is between 6-23, e.g., 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22 or 23. In some embodiments, the CDR3 length is at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, or at least 23.
Transferrin receptor 1 (TFR1)
TFR1, also known as cluster of differentiation 71 (CD71) , is widely expressed and can bind to transferrin (Tf) with high affinity. Human TFR1 is a 90 kDa type II transmembrane glycoprotein consisting of 760 amino acids that is found as a dimer (180 kDa) linked by disulfide bonds on the cell surface. The TFR1 monomer is composed of a large extracellular, C-terminal domain of 671 amino acids containing the Tf-binding site, a transmembrane domain (28 amino acids) , and an intracellular N-terminal domain (61 amino acids) . The C-terminal extracellular domain contains three N-linked glycosylation sites at asparagine residues 251, 317, and 727 and one O-linked glycosylation site at threonine 104, which are all required for adequate function of the receptor.
Transferrin (Tf) It is an 80 kDa glycoprotein composed of two 40 kDa subunits, known as the N-and C-lobes that are separated by a short linker sequence. Each subunit is capable of binding to one free ferric iron (Fe3+) and thus, Tf may have up to two atoms of iron attached. Tf in its iron free form, apo-Tf, binds Fe3+ with high efficiency in the blood and transports it to the cell surface for internalization through the interaction with TFR1. As a membrane protein regulating iron import, TFR1 is a member of the TFR family that shows nanomolar affinity to transferrin (Tf) bound to Fe3+. The complex of Tf-TFR1 is internalized through endocytosis
mediated by clathrin, and Fe3+ is disassociated from Tf when pH decreases to 5.5. At this pH, apo-Tf and TFR1 are still associated and recycled to cell surface with physiological pH, so the former is released.
Iron uptake by transferrin receptor is an important way for cancer cells to absorb iron, thus accumulating evidence has proven that TFR1 participated in tumor onset and progression, and its expression was dysregulated significantly in many cancers. The relationship between TFR1 and cancers has been revealed, rendering TFR1 a valuable pharmaceutical target for intervening with cancers.
TFR1 expressed on the endothelial cells of the blood-brain barrier is used also in preclinical research to allow the delivery of large molecules including antibodies into the brain. The TFR1 targeting antibodies can cross the blood-brain barrier, without interfering with the uptake of iron.
A detailed description of TFR1, Tf, and their functions can be found, e.g., in Candelaria, P.V., et al. "Antibodies targeting the transferrin receptor 1 (TfR1) as direct anti-cancer agents. " Frontiers in Immunology 12 (2021) : 607692; and Shen, Y., et al. "Transferrin receptor 1 in cancer: a new sight for cancer therapy. " American Journal of Cancer Research 8.6 (2018) : 916; each of which is incorporated by reference in its entirety.
Heavy chain single variable domain (VHH) antibodies
Monoclonal and recombinant antibodies are important tools in medicine and biotechnology. Like all mammals, camelids (e.g., llamas) can produce conventional antibodies made of two heavy chains and two light chains bound together with disulfide bonds in a Y shape (e.g., IgG1) . However, they also produce two unique subclasses of IgG: IgG2 and IgG3, also known as heavy chain IgG. These antibodies are made of only two heavy chains, which lack the CH1 region but still bear an antigen-binding domain at their N-terminus called VHH (or nanobody) . Conventional Ig require the association of variable regions from both heavy and light chains to allow a high diversity of antigen-antibody interactions. Although isolated heavy and light chains still show this capacity, they exhibit very low affinity when compared to paired heavy and light chains. The unique feature of heavy chain IgG is the capacity of their monomeric antigen binding regions to bind antigens with specificity, affinity and especially diversity that are comparable to conventional antibodies without the need of pairing with another region. This feature is mainly due to a couple of major variations within the amino acid sequence of the variable region of the two heavy chains, which induce deep conformational changes when compared to conventional Ig. Major substitutions in the variable regions prevent the light chains from binding to the heavy chains, but also prevent unbound heavy chains from being recycled by the immunoglobulin binding proteins.
The single variable domain of these antibodies (designated VHH, sdAb, or nanobody) is the smallest antigen-binding domain generated by adaptive immune systems. The third Complementarity Determining Region (CDR3) of the variable region of these antibodies has been found to be twice as long as the conventional ones. This results in an increased interaction surface with the antigen as well as an increased diversity of antigen-antibody interactions, which compensates the absence of the light chains. With a long complementarity-determining region 3 (CDR3) , VHHs can extend into crevices on proteins that are not accessible to conventional antibodies, including functionally interesting sites such as the active site of an enzyme or the receptor-binding canyon on a virus surface.
VHHs offer numerous other advantages compared to conventional antibodies carrying variable domains (VH and VL) of conventional antibodies, including higher stability, solubility, expression yields, and refolding capacity, as well as better in vivo tissue penetration and internalization. Moreover, in contrast to the VH domains of conventional antibodies, VHH do not display an intrinsic tendency to bind to light chains. Since VHH do not bind to VL domains, it is much easier to reformat VHHs into multi-specific (e.g., bispecific antibody) constructs than constructs containing conventional VH-VL pairs or single domains based on VH domains.
The disclosure provides e.g., anti-TFR1 antibodies, the modified antibodies thereof, the chimeric antibodies thereof, and the humanized antibodies thereof.
The CDR sequences for 23B8, and 23B8 derived antibodies (e.g., humanized antibodies) include CDRs of the VHH domain as set forth in SEQ ID NOs: 1, 2, and 3, respectively, as defined by Kabat numbering. The CDRs can also be defined by IMGT system. Under the IMGT numbering, the CDRs of the VHH domain are set forth in SEQ ID NOs: 13, 14, and 15, respectively.
The CDR sequences for 24A1, and 24A1 derived antibodies (e.g., humanized antibodies) include CDRs of the VHH domain as set forth in SEQ ID NOs: 4, 5, and 6, respectively, as defined by Kabat numbering. The CDRs can also be defined by IMGT system. Under the IMGT numbering, the CDRs of the VHH domain are set forth in SEQ ID NOs: 16, 17, and 18, respectively.
The CDR sequences for 24C9, and 24C9 derived antibodies (e.g., humanized antibodies) include CDRs of the VHH domain as set forth in SEQ ID NOs: 7, 8, and 9, respectively, as defined by Kabat numbering. The CDRs can also be defined by IMGT system. Under the IMGT numbering, the CDRs of the VHH domain are set forth in SEQ ID NOs: 19, 20, and 21, respectively.
The CDR sequences for 24G5, and 24G5 derived antibodies (e.g., humanized antibodies) include CDRs of the VHH domain as set forth in SEQ ID NOs: 10, 11, and 12, respectively, as defined by Kabat numbering. The CDRs can also be defined by IMGT system. Under the IMGT numbering, the CDRs of the VHH domain are set forth in SEQ ID NOs: 22, 23, and 24, respectively.
The amino acid sequence for the VHH domain of 23B8 antibody is set forth in SEQ ID NO: 25. The amino acid sequence for the VHH domain of 24A1 antibody is set forth in SEQ ID NO: 26. The amino acid sequence for the VHH domain of 24C9 antibody is set forth in SEQ ID NO: 27. The amino acid sequence for the VHH domain of 24G5 antibody is set forth in SEQ ID NO: 28.
The amino acid sequences for various modified or humanized VHH are also provided. As there are different ways to modify or humanize a heavy-chain antibody (e.g., a sequence can be modified with different amino acid substitutions) , the VHH domain of a heavy-chain antibody can have more than one version of humanized sequences. In some embodiments, the humanized VHH domain is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%identical to any sequence of SEQ ID NOS: 25-28.
Furthermore, in some embodiments, the antibodies or antigen-binding fragments thereof described herein can also contain one, two, or three VHH domain CDRs selected from the group of SEQ ID NOs: 1-3, SEQ ID NOs: 4-6, SEQ ID NOs: 7-9, SEQ ID NOs: 10-12, SEQ ID NOs: 13-15, SEQ ID NOs: 16-18, SEQ ID NOs: 19-21, and SEQ ID NOs: 22-24.
In some embodiments, the antibodies can have a heavy chain single variable domain (VHH) comprising complementarity determining regions (CDRs) 1, 2, 3, wherein the CDR1
region comprises or consists of an amino acid sequence that is at least 80%, 85%, 90%, or 95%identical to a selected VHH CDR1 amino acid sequence, the CDR2 region comprises or consists of an amino acid sequence that is at least 80%, 85%, 90%, or 95%identical to a selected VHH CDR2 amino acid sequence, and the CDR3 region comprises or consists of an amino acid sequence that is at least 80%, 85%, 90%, or 95%identical to a selected VHH CDR3 amino acid sequence. The selected VHH CDRs 1, 2, 3 amino acid sequences are shown in FIG. 1 and FIG. 2.
In some embodiments, the antibody or an antigen-binding fragment described herein can contain a heavy chain single variable domain (VHH) containing one, two, or three of VHH CDR1 with zero, one or two amino acid insertions, deletions, or substitutions; VHH CDR2 with zero, one or two amino acid insertions, deletions, or substitutions; VHH CDR3 with zero, one or two amino acid insertions, deletions, or substitutions, wherein VHH CDR1, VHH CDR2, and VHH CDR3 are selected from the CDRs in FIG. 3.
In some embodiments, the antibody or an antigen-binding fragment described herein can contain a heavy chain single variable domain (VHH) containing one, two, or three of the CDRs of SEQ ID NO: 1 with zero, one or two amino acid insertions, deletions, or substitutions; SEQ ID NO: 2 with zero, one or two amino acid insertions, deletions, or substitutions; SEQ ID NO: 3 with zero, one or two amino acid insertions, deletions, or substitutions.
In some embodiments, the antibody or an antigen-binding fragment described herein can contain a heavy chain single variable domain (VHH) containing one, two, or three of the CDRs of SEQ ID NO: 4 with zero, one or two amino acid insertions, deletions, or substitutions; SEQ ID NO: 5 with zero, one or two amino acid insertions, deletions, or substitutions; SEQ ID NO: 6 with zero, one or two amino acid insertions, deletions, or substitutions.
In some embodiments, the antibody or an antigen-binding fragment described herein can contain a heavy chain single variable domain (VHH) containing one, two, or three of the CDRs of SEQ ID NO: 7 with zero, one or two amino acid insertions, deletions, or substitutions; SEQ ID NO: 8 with zero, one or two amino acid insertions, deletions, or substitutions; SEQ ID NO: 9 with zero, one or two amino acid insertions, deletions, or substitutions.
In some embodiments, the antibody or an antigen-binding fragment described herein can contain a heavy chain single variable domain (VHH) containing one, two, or three of the CDRs of SEQ ID NO: 10 with zero, one or two amino acid insertions, deletions, or substitutions; SEQ ID NO: 11 with zero, one or two amino acid insertions, deletions, or substitutions; SEQ ID NO: 12 with zero, one or two amino acid insertions, deletions, or substitutions.
In some embodiments, the antibody or an antigen-binding fragment described herein can contain a heavy chain single variable domain (VHH) containing one, two, or three of the CDRs of SEQ ID NO: 13 with zero, one or two amino acid insertions, deletions, or substitutions; SEQ ID NO: 14 with zero, one or two amino acid insertions, deletions, or substitutions; SEQ ID NO: 15 with zero, one or two amino acid insertions, deletions, or substitutions.
In some embodiments, the antibody or an antigen-binding fragment described herein can contain a heavy chain single variable domain (VHH) containing one, two, or three of the CDRs of SEQ ID NO: 16 with zero, one or two amino acid insertions, deletions, or substitutions; SEQ ID NO: 17 with zero, one or two amino acid insertions, deletions, or substitutions; SEQ ID NO: 18 with zero, one or two amino acid insertions, deletions, or substitutions.
In some embodiments, the antibody or an antigen-binding fragment described herein can contain a heavy chain single variable domain (VHH) containing one, two, or three of the CDRs of SEQ ID NO: 19 with zero, one or two amino acid insertions, deletions, or substitutions; SEQ
ID NO: 20 with zero, one or two amino acid insertions, deletions, or substitutions; SEQ ID NO: 21 with zero, one or two amino acid insertions, deletions, or substitutions.
In some embodiments, the antibody or an antigen-binding fragment described herein can contain a heavy chain single variable domain (VHH) containing one, two, or three of the CDRs of SEQ ID NO: 22 with zero, one or two amino acid insertions, deletions, or substitutions; SEQ ID NO: 23 with zero, one or two amino acid insertions, deletions, or substitutions; SEQ ID NO: 24 with zero, one or two amino acid insertions, deletions, or substitutions.
The insertions, deletions, and substitutions can be within the CDR sequence, or at one or both terminal ends of the CDR sequence. In some embodiments, the CDR is determined based on Kabat numbering scheme. In some embodiments, the CDR is determined based on Chothia numbering scheme. In some embodiments, the CDR is determined based on a combination numbering scheme. In some embodiments, the CDR is determined based on IMGT numbering scheme.
The disclosure also provides antibodies or antigen-binding fragments thereof that bind to TFR1 (human TFR1) . The antibodies or antigen-binding fragments thereof contain a heavy chain single variable region (VHH) comprising or consisting of an amino acid sequence that is at least 80%, 85%, 90%, or 95%identical to a selected VHH sequence. In some embodiments, the selected VHH sequence is SEQ ID NO: 25. In some embodiments, the selected VHH sequence is SEQ ID NO: 26. In some embodiments, the selected VHH sequence is SEQ ID NO: 27. In some embodiments, the selected VHH sequence is SEQ ID NO: 28.
To determine the percent identity of two amino acid sequences, or of two nucleic acid sequences, the sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in one or both of a first and a second amino acid or nucleic acid sequence for optimal alignment and non-homologous sequences can be disregarded for comparison purposes) . The amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared. When a position in the first sequence is occupied by the same amino acid residue or nucleotide as the corresponding position in the second sequence, then the molecules are identical at that position. The percent identity between the two sequences is a function of the number of identical positions shared by the sequences, taking into account the number of gaps, and the length of each gap, which need to be introduced for optimal alignment of the two sequences. For purposes of illustration, the comparison of sequences and determination of percent identity between two sequences can be accomplished, e.g., using a Blossum 62 scoring matrix with a gap penalty of 12, a gap extend penalty of 4, and a frameshift gap penalty of 5.
The disclosure also provides nucleic acid comprising a polynucleotide encoding a polypeptide comprising an immunoglobulin heavy chain single variable domain (VHH) . The VHH comprises CDRs as shown in FIG. 1 and FIG. 2, or has sequences as shown in FIG. 3.
The antibodies and antigen-binding fragments can also be antibody variants (including derivatives and conjugates) of antibodies or antibody fragments and multi-specific (e.g., bi-specific) antibodies or antibody fragments. Additional antibodies provided herein are polyclonal, monoclonal, multi-specific (multimeric, e.g., bi-specific) , human antibodies, chimeric antibodies (e.g., human-mouse chimera) , single-chain antibodies, intracellularly-made antibodies (i.e., intrabodies) , and antigen-binding fragments thereof.
In some embodiments, the antibodies or antigen-binding fragments thereof comprises an Fc domain that can be originated from various types (e.g., IgG, IgE, IgM, IgD, IgA, and IgY) , class (e.g., IgG1, IgG2, IgG3, IgG4, IgA1, and IgA2) , or subclass. In some embodiments, the Fc domain is originated from an IgG antibody or antigen-binding fragment thereof. In some
embodiments, the Fc domain comprises one, two, three, four, or more heavy chain constant regions.
The present disclosure also provides an antibody or antigen-binding fragment thereof that cross-competes with any antibody or antigen-binding fragment as described herein. The cross-competing assay is known in the art, and is described e.g., in Moore et al., "Antibody cross-competition analysis of the human immunodeficiency virus type 1 gp120 exterior envelope glycoprotein. " Journal of virology 70.3 (1996) : 1863-1872, which is incorporated herein reference in its entirety. In one aspect, the present disclosure also provides an antibody or antigen-binding fragment thereof that binds to the same epitope or region as any antibody or antigen-binding fragment as described herein. The epitope binning assay is known in the art, and is described e.g., in Estep et al. "High throughput solution-based measurement of antibody-antigen affinity and epitope binning. " MAbs. Vol. 5. No. 2. Taylor &Francis, 2013, which is incorporated herein reference in its entirety.
In some embodiments, the antibody or antigen-binding fragment thereof comprises a heavy chain single variable domain (VHH) CDR1 selected from SEQ ID NOs: 1, 4, 7, 10, 13, 16, 19, and 22.
In some embodiments, the antibody or antigen-binding fragment thereof comprises a heavy chain single variable domain (VHH) CDR2 selected from SEQ ID NOs: 2, 5, 8, 11, 14, 17, 20, and 23.
In some embodiments, the antibody or antigen-binding fragment thereof comprises a heavy chain single variable domain (VHH) CDR3 selected from SEQ ID NOs: 3, 6, 9, 12, 15, 18, 21, and 24.
Antibody characteristics
TFR1 performs a critical role in cellular iron uptake through the interaction with iron-bound TF. Iron is required for multiple cellular processes and is essential for DNA synthesis and, thus, cellular proliferation. Due to its central role in cancer cell pathology, malignant cells often overexpress TFR1 and this increased expression can be associated with poor prognosis in different types of cancer. The elevated levels of TfR1 expression on malignant cells, together with its extracellular accessibility, ability to internalize, and central role in cancer cell pathology make this receptor an attractive target for antibody-mediated therapy.
In some embodiments, the antibodies or antigen-binding fragments thereof described herein cannot block the binding between TFR1 and TF. In some embodiments, the antibodies or antigen-binding fragments thereof described herein can block the binding between TFR1 and TF. In some embodiments, the antibodies or antigen-binding fragments thereof described herein can be conjugated to anti-cancer agents that are internalized by receptor-mediated endocytosis. In some embodiments, the antibodies or antigen-binding fragments thereof described herein can disrupt the function of the receptor. In some embodiments, the antibodies or antigen-binding fragments thereof described herein cannot induce Fc effector functions, therefore preventing or ameliorating their negative effects to normal cells.
The disclosure provides antibodies or antigen-binding fragments thereof comprising a human Fc domain, which induce Fc-dependent effector functions by at least or about at least or about 1 fold, at least or about 2 folds, at least or about 3 folds, at least or about 4 folds, at least or about 5 folds, at least or about 6 folds, at least or about 7 folds, at least or about 8 folds, at least or about 9 folds, at least or about 10 folds, at least or about 20 folds, at least or about 30 folds, at
least or about 40 folds, at least or about 50 folds, or at least or about 100 folds as compared when no antibodies or antigen-binding fragments thereof as described herein are present.
The disclosure provides antibodies or antigen-binding fragments thereof comprising a human Fc domain, which induce host immune response by at least or about at least or about 1 fold, at least or about 2 folds, at least or about 3 folds, at least or about 4 folds, at least or about 5 folds, at least or about 6 folds, at least or about 7 folds, at least or about 8 folds, at least or about 9 folds, at least or about 10 folds, at least or about 20 folds, at least or about 30 folds, at least or about 40 folds, at least or about 50 folds, or at least or about 100 folds as compared when no antibodies or antigen-binding fragments thereof as described herein are present.
The disclosure provides antibodies or antigen-binding fragments thereof that can internalize into human brain cells (e.g., cortical microvascular endothelial cells) that the endocytosis rate is at least 50%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%. In some embodiments, the endocytosis rate of the antibodies or antigen-binding fragments thereof described herein is at least 1-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 50-fold, 100-fold, 500-fold, or 1000-fold as compared to that of an isotype control antibody.
In some embodiments, provided herein is an antibody or antigen-binding fragment thereof comprising a single heavy chain. In some embodiments, provided herein is an antibody or antigen-binding fragment thereof comprising a pair of heavy chains. In some embodiments, the heavy chain pair is linked by disulfide bonds. In some embodiments, the heavy chain pair comprises knob-in-hole modifications. In some embodiments, the heavy chain comprises a human IgG Fc domain. In some embodiments, the antibody or antigen-binding fragment thereof comprises in each heavy chain at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 VHH domains. In some embodiments, the VHH domains in each heavy chain specifically bind to the same epitope. In some embodiments, the VHH domains in each heavy chain specifically bind to different epitopes. In some embodiments, the VHH domains in each heavy chain bind to at least 1, 2, 3, 4, or 5 different epitopes.
In some embodiments, the antibody or antigen-binding fragment thereof is a bi-specific antibody, or tri-specific antibody. In some embodiments, the antibody or antigen-binding fragment thereof can specifically bind to at least 4, 5, or 6 antigens.
In some embodiments, the antibody (or antigen-binding fragment thereof) specifically binds to TFR1 with a dissociation rate (koff) of less than 0.1 s-1, less than 0.01 s-1, less than 0.001 s-1, less than 0.0001 s-1, or less than 0.00001 s-1. In some embodiments, the dissociation rate (koff) is greater than 0.01 s-1, greater than 0.001 s-1, greater than 0.0001 s-1, greater than 0.00001 s-1, or greater than 0.000001 s-1.
In some embodiments, kinetic association rates (kon) is greater than 1 × 102/Ms, greater than 1 × 103/Ms, greater than 1 × 104/Ms, greater than 1 × 105/Ms, or greater than 1 × 106/Ms. In some embodiments, kinetic association rates (kon) is less than 1 × 105/Ms, less than 1 × 106/Ms, or less than 1 × 107/Ms.
Affinities can be deduced from the quotient of the kinetic rate constants (KD=koff/kon) . In some embodiments, KD is less than 1 × 10-6 M, less than 1 × 10-7 M, less than 1 × 10-8 M, less than 1 × 10-9 M, or less than 1 × 10-10 M. In some embodiments, the KD is less than 50 nM, 30 nM, 20 nM, 15 nM, 10 nM, 9 nM, 8 nM, 7 nM, 6 nM, 5 nM, 4 nM, 3 nM, 2 nM, or 1 nM. In some embodiments, KD is greater than 1 x 10-7 M, greater than 1 x 10-8 M, greater than 1 x 10-9 M, greater than 1 x 10-10 M, greater than 1 x 10-11 M, or greater than 1 x 10-12 M.
General techniques for measuring the affinity of an antibody for an antigen include, e.g., ELISA, RIA, and surface plasmon resonance (SPR) . In some embodiments, the antibody binds to human TFR1, monkey TFR1, mouse TFR1, or chimeric TFR1. In some embodiments, the antibody does not bind to human TFR1, monkey TFR1, mouse TFR1, or chimeric TFR1.
In some embodiments, thermal stabilities are determined. The antibodies or antigen-binding fragments as described herein can have a Tm (melting temperature) greater than 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, or 95 ℃. In some embodiments, Tm is less than 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, or 95 ℃. The antibodies or antigen-binding fragments as described herein can have a Tagg (aggregation temperature, e.g., Tagg at 266 nm (Tagg266) or Tagg at 473 nm (Tagg473) ) great than 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, or 95 ℃. In some embodiments, Tagg is less than 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, or 95 ℃.
In some embodiments, the Fc region is human IgG1, human IgG2, human IgG3, or human IgG4.
In some embodiments, the antibodies or antigen binding fragments thereof have a functional Fc region. In some embodiments, the antibodies or antigen binding fragments thereof comprise a human IgG1 Fc region. In some embodiments, the human IgG1 Fc region comprises an amino acid sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identical to SEQ ID NO: 34.
In some embodiments, the antibodies or antigen binding fragments do not have an Fc region. For example, the antibody (or antigen-binding fragment thereof) is a polypeptide comprising one or more VHH domains that are interconnected by linker peptides. In some embodiments, the antibody comprises at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 VHH domain. In some embodiments, the VHH domains specifically bind to the same epitope. In some embodiments, the VHH domains bind to different epitopes. In some embodiments, the VHH domains bind to at least 1, 2, 3, 4, or 5 different epitopes.
In some embodiments, the antibodies or antigen binding fragments thereof do not have a functional Fc region. In some embodiments, the Fc region has LALA mutations (L234A and L235A mutations in EU numbering) , or LALA-PG mutations (L234A, L235A, P329G mutations in EU numbering) . In some embodiments, the Fc region has a mutation at position 297 (e.g., N297A) according to EU numbering. In some embodiments, the mutated human IgG1 Fc region comprises an amino acid sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identical to SEQ ID NO: 35.
In some embodiments, after being administered to a subject after 6 hours, 12 hours, 24 hours, 36 hours, 48 hours, 60 hours, or 72 hours, concentration of the antibody or antigen binding fragment thereof described herein in brain (e.g., whole brain or parenchyma) can be greater than 10%, 20%, 30%, 40%, 50%, 60%, 70%, or 80%of its concentration immediately (e.g., 0.5 hour) after administration. In some embodiments, after being administered to a subject after 6 hours, 12 hours, 24 hours, 36 hours, 48 hours, 60 hours, or 72 hours, concentration of the antibody or antigen binding fragment thereof described herein in brain (e.g., whole brain or parenchyma) can be at least 1-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 20-fold, 30-fold, 40-fold, 50-fold, 60-fold, 70-fold, 80-fold, 90-fold, 100-fold, 200-fold,
300-fold, 400-fold, 500-fold, 1000-fold, 2000-fold, 5000-fold, or 10000-fold of the concentration of a control antibody (e.g., hIgG1 or JR141-N) , or the concentration in serum of the subject.
Methods of making anti-TFR1 antibodies
Variants of the antibodies or antigen-binding fragments described herein can be prepared by introducing appropriate nucleotide changes into the DNA encoding a human, humanized, or chimeric antibody, or antigen-binding fragment thereof described herein, or by peptide synthesis. Such variants include, for example, deletions, insertions, or substitutions of residues within the amino acids sequences that make-up the antigen-binding site of the antibody or an antigen-binding domain. In a population of such variants, some antibodies or antigen-binding fragments will have increased affinity for the target protein, e.g., TFR1. Any combination of deletions, insertions, and/or combinations can be made to arrive at an antibody or antigen-binding fragment thereof that has increased binding affinity for the target. The amino acid changes introduced into the antibody or antigen-binding fragment can also alter or introduce new post-translational modifications into the antibody or antigen-binding fragment, such as changing (e.g., increasing or decreasing) the number of glycosylation sites, changing the type of glycosylation site (e.g., changing the amino acid sequence such that a different sugar is attached by enzymes present in a cell) , or introducing new glycosylation sites. In some embodiments, the heavy-chain antibody or antigen-binding fragment thereof described herein is obtained by immunizing any of the genetically modified animals (e.g., mice with complete human heavy chain variable domain substitution in situ, combined with a modified constant region) described e.g., in PCT/CN2022/119188.
Humanized antibodies include antibodies having variable and constant regions derived from (or having the same amino acid sequence as those derived from) human germline immunoglobulin sequences of human immunoglobulin scaffold sequences. Humanized antibodies may include amino acid residues not encoded by human germline immunoglobulin sequences (e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo) . Accordingly, “humanized” antibodies are chimeric antibodies wherein sequences from a non-human species are substituted by the corresponding human sequences.
Ordinarily, amino acid sequence variants of the human, humanized, or chimeric anti-TFR1 antibody will contain an amino acid sequence having at least 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%percent identity with a sequence present in the VHH domain of the original antibody.
Identity or homology with respect to an original sequence is usually the percentage of amino acid residues present within the candidate sequence that are identical with a sequence present within the human, humanized, or chimeric anti-TFR1 antibody or fragment, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity.
Additional modifications to the anti-TFR1 antibodies or antigen-binding fragments can be made. For example, a cysteine residue (s) can be introduced into the Fc region, thereby allowing interchain disulfide bond formation in this region. The homodimeric antibody thus generated may have any increased half-life in vitro and/or in vivo. Homodimeric antibodies with increased half-life in vitro and/or in vivo can also be prepared using heterobifunctional cross-linkers as described, for example, in Wolff et al. Wolff et al. ( "Monoclonal antibody homodimers:
enhanced antitumor activity in nude mice. " Cancer research 53.11 (1993) : 2560-2565) . Alternatively, an antibody can be engineered which has dual Fc regions.
In some embodiments, a covalent modification can be made to the anti-TFR1 antibody or antigen-binding fragment thereof. These covalent modifications can be made by chemical or enzymatic synthesis, or by enzymatic or chemical cleavage. Other types of covalent modifications of the antibody or antibody fragment are introduced into the molecule by reacting targeted amino acid residues of the antibody or fragment with an organic derivatization agent that is capable of reacting with selected side chains or the N-or C-terminal residues.
In some embodiments, antibody variants are provided having a carbohydrate structure that lacks fucose attached (directly or indirectly) to an Fc region. For example, the amount of fucose in such antibody composition may be from 1%to 80%, from 1%to 65%, from 5%to 65%or from 20%to 40%. The amount of fucose is determined by calculating the average amount of fucose within the sugar chain at Asn297, relative to the sum of all glycostructures attached to Asn 297 (e.g. complex, hybrid and high mannose structures) as measured by MALDI-TOF mass spectrometry, as described in WO 2008/077546, for example. Asn297 refers to the asparagine residue located at about position 297 in the Fc region (Eu numbering of Fc region residues; or position 314 in Kabat numbering) ; however, Asn297 may also be located about ±3 amino acids upstream or downstream of position 297, i.e., between positions 294 and 300, due to minor sequence variations in antibodies. Such fucosylation variants may have improved ADCC function. In some embodiments, to reduce glycan heterogeneity, the Fc region of the antibody can be further engineered to replace the Asparagine at position 297 with Alanine (N297A) .
The present disclosure also provides recombinant vectors (e.g., an expression vectors) that include an isolated polynucleotide disclosed herein (e.g., a polynucleotide that encodes a polypeptide disclosed herein) , host cells into which are introduced the recombinant vectors (i.e., such that the host cells contain the polynucleotide and/or a vector comprising the polynucleotide) , and the production of recombinant antibody polypeptides or fragments thereof by recombinant techniques.
As used herein, a “vector” is any construct capable of delivering one or more polynucleotide (s) of interest to a host cell when the vector is introduced to the host cell. An “expression vector” is capable of delivering and expressing the one or more polynucleotide (s) of interest as an encoded polypeptide in a host cell into which the expression vector has been introduced. Thus, in an expression vector, the polynucleotide of interest is positioned for expression in the vector by being operably linked with regulatory elements such as a promoter, enhancer, and/or a poly-Atail, either within the vector or in the genome of the host cell at or near or flanking the integration site of the polynucleotide of interest such that the polynucleotide of interest will be translated in the host cell introduced with the expression vector.
A vector can be introduced into the host cell by methods known in the art, e.g., electroporation, chemical transfection (e.g., DEAE-dextran) , transformation, transfection, and infection and/or transduction (e.g., with recombinant virus) . Thus, non-limiting examples of vectors include viral vectors (which can be used to generate recombinant virus) , naked DNA or RNA, plasmids, cosmids, phage vectors, and DNA or RNA expression vectors associated with cationic condensing agents.
In some implementations, a polynucleotide disclosed herein (e.g., a polynucleotide that encodes a polypeptide disclosed herein) is introduced using a viral expression system (e.g., vaccinia or other pox virus, retrovirus, or adenovirus) , which may involve the use of a non-pathogenic (defective) , replication competent virus, or may use a replication defective virus. In
the latter case, viral propagation generally will occur only in complementing virus packaging cells. Suitable systems are disclosed, for example, in Fisher-Hoch et al., 1989, Proc. Natl. Acad. Sci. USA 86: 317-321; Flexner et al., 1989, Ann. N. Y. Acad Sci. 569: 86-103; Flexner et al., 1990, Vaccine, 8: 17-21; U.S. Pat. Nos. 4,603,112, 4,769,330, and 5,017,487; WO 89/01973; U.S. Pat. No. 4,777,127; GB 2,200,651; EP 0,345,242; WO 91/02805; Berkner-Biotechniques, 6: 616-627, 1988; Rosenfeld et al., 1991, Science, 252: 431-434; Kolls et al., 1994, Proc. Natl. Acad. Sci. USA, 91: 215-219; Kass-Eisler et al., 1993, Proc. Natl. Acad. Sci. USA, 90: 11498-11502; Guzman et al., 1993, Circulation, 88: 2838-2848; and Guzman et al., 1993, Cir. Res., 73: 1202-1207. Techniques for incorporating DNA into such expression systems are well known to those of ordinary skill in the art. The DNA may also be “naked, ” as described, for example, in Ulmer et al., 1993, Science, 259: 1745-1749, and Cohen, 1993, Science, 259: 1691-1692. The uptake of naked DNA may be increased by coating the DNA onto biodegradable beads that are efficiently transported into the cells.
For expression, the DNA insert comprising an antibody-encoding or polypeptide-encoding polynucleotide disclosed herein can be operatively linked to an appropriate promoter (e.g., a heterologous promoter) , such as the phage lambda PL promoter, the E. coli lac, trp and tac promoters, the SV40 early and late promoters and promoters of retroviral LTRs, to name a few. Other suitable promoters are known to the skilled artisan. In some embodiments, the promoter is a cytomegalovirus (CMV) promoter. The expression constructs can further contain sites for transcription initiation, termination and, in the transcribed region, a ribosome binding site for translation. The coding portion of the mature transcripts expressed by the constructs may include a translation initiating at the beginning and a termination codon (UAA, UGA, or UAG) appropriately positioned at the end of the polypeptide to be translated.
As indicated, the expression vectors can include at least one selectable marker. Such markers include dihydrofolate reductase or neomycin resistance for eukaryotic cell culture and tetracycline or ampicillin resistance genes for culturing in E. coli and other bacteria. Representative examples of appropriate hosts include, but are not limited to, bacterial cells, such as E. coli, Streptomyces, and Salmonella typhimurium cells; fungal cells, such as yeast cells; insect cells such as Drosophila S2 and Spodoptera Sf9 cells; animal cells such as CHO, COS, Bowes melanoma, and HK 293 cells; and plant cells. Appropriate culture mediums and conditions for the host cells described herein are known in the art.
Non-limiting vectors for use in bacteria include pQE70, pQE60 and pQE-9, available from Qiagen; pBS vectors, Phagescript vectors, Bluescript vectors, pNH8A, pNH16a, pNH18A, pNH46A, available from Stratagene; and ptrc99a, pKK223-3, pKK233-3, pDR540, pRIT5 available from Pharmacia. Non-limiting eukaryotic vectors include pWLNEO, pSV2CAT, pOG44, pXT1 and pSG available from Stratagene; and pSVK3, pBPV, pMSG and pSVL available from Pharmacia. Other suitable vectors will be readily apparent to the skilled artisan.
Non-limiting bacterial promoters suitable for use include the E. coli lacI and lacZ promoters, the T3 and T7 promoters, the gpt promoter, the lambda PR and PL promoters and the trp promoter. Suitable eukaryotic promoters include the CMV immediate early promoter, the HSV thymidine kinase promoter, the early and late SV40 promoters, the promoters of retroviral LTRs, such as those of the Rous sarcoma virus (RSV) , and metallothionein promoters, such as the mouse metallothionein-I promoter.
In the yeast Saccharomyces cerevisiae, a number of vectors containing constitutive or inducible promoters such as alpha factor, alcohol oxidase, and PGH can be used.
Introduction of the construct into the host cell can be affected by calcium phosphate transfection, DEAE-dextran mediated transfection, cationic lipid-mediated transfection, electroporation, transduction, infection or other methods. Such methods are described in many standard laboratory manuals, such as Davis et al., Basic Methods in Molecular Biology (1986) , which is incorporated herein by reference in its entirety.
Transcription of DNA encoding an antibody of the present disclosure by higher eukaryotes may be increased by inserting an enhancer sequence into the vector. Enhancers are cis-acting elements of DNA, usually about from 10 to 300 bp that act to increase transcriptional activity of a promoter in a given host cell-type. Examples of enhancers include the SV40 enhancer, which is located on the late side of the replication origin at base pairs 100 to 270, the cytomegalovirus early promoter enhancer, the polyoma enhancer on the late side of the replication origin, and adenovirus enhancers.
For secretion of the translated protein into the lumen of the endoplasmic reticulum, into the periplasmic space or into the extracellular environment, appropriate secretion signals may be incorporated into the expressed polypeptide. The signals may be endogenous to the polypeptide or they may be heterologous signals.
The polypeptide (e.g., antibody) can be expressed in a modified form, such as a fusion protein (e.g., a GST-fusion) or with a histidine-tag, and may include not only secretion signals, but also additional heterologous functional regions. For instance, a region of additional amino acids, particularly charged amino acids, may be added to the N-terminus of the polypeptide to improve stability and persistence in the host cell, during purification, or during subsequent handling and storage. Also, peptide moieties can be added to the polypeptide to facilitate purification. Such regions can be removed prior to final preparation of the polypeptide. The addition of peptide moieties to polypeptides to engender secretion or excretion, to improve stability and to facilitate purification, among others, are familiar and routine techniques in the art.
The disclosure also provides a nucleic acid sequence that is at least 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%identical to any nucleotide sequence as described herein, and an amino acid sequence that is at least 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%identical to any amino acid sequence as described herein. In some embodiments, the disclosure relates to nucleotide sequences encoding any peptides that are described herein, or any amino acid sequences that are encoded by any nucleotide sequences as described herein. In some embodiments, the nucleic acid sequence is less than 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 150, 200, 250, 300, 350, 400, 500, or 600 nucleotides. In some embodiments, the amino acid sequence is less than 5, 6, 7, 8, 9, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, or 200 amino acid residues.
In some embodiments, the amino acid sequence (i) comprises an amino acid sequence; or (ii) consists of an amino acid sequence, wherein the amino acid sequence is any one of the sequences as described herein.
In some embodiments, the nucleic acid sequence (i) comprises a nucleic acid sequence; or (ii) consists of a nucleic acid sequence, wherein the nucleic acid sequence is any one of the sequences as described herein.
In some embodiments, the antibody or antigen-binding fragment thereof is expressed in yeast, insect cells, or mammalian cells (e.g., CHO cells) .
Methods of treatment and diagnosis
The anti-TFR1 antibodies or antibody or antigen-binding fragments thereof of the present disclosure can be used for various therapeutic purposes. In one aspect, the disclosure provides methods for treating a brain disease (e.g., brain cancer, dementia, or Alzheimer’s disease) in a subject, methods of identifying a subject having a brain disease (e.g., brain cancer, dementia, or Alzheimer’s disease) , methods of reducing the risk of developing a brain disease, or methods of reducing the risk of developing additional symptoms in a subject. In some embodiments, the treatment can halt, slow, retard, or inhibit progression of a brain disease (e.g., brain cancer, dementia, or Alzheimer’s disease) . In some embodiments, the treatment can result in the reduction of in the number, severity, and/or duration of one or more symptoms of a brain disease (e.g., brain cancer, dementia, or Alzheimer’s disease) in a subject.
In one aspect, the disclosure features methods that include administering a therapeutically effective amount of an antibody or antigen-binding fragment thereof disclosed herein to a subject in need thereof (e.g., a subject having, or identified or diagnosed as having, a brain disease) .
In one aspect, the disclosure features methods that carry a therapeutic agent to cross the blood brain barrier. In some embodiments, the antibodies or antigen-binding fragments thereof as described herein are linked to the therapeutic agent. In some embodiments, the therapeutic agent is an antibody, an antigen binding fragment thereof, a small molecule, or an antibody-drug conjugate.
In some embodiments, the compositions and methods disclosed herein can be used for treatment of patients at risk for a brain disease (e.g., brain cancer, dementia, or Alzheimer’s disease) . Patients with a brain disease (e.g., brain cancer, dementia, or Alzheimer’s disease) can be identified with various methods known in the art.
In some embodiments, the brain disease is a brain cancer.
In one aspect, the disclosure is related to methods of decreasing the rate of tumor growth, including contacting a tumor cell with an effective amount of a composition including the antibody or antigen-binding fragment thereof, or the antibody-drug conjugate described herein. In one aspect, the disclosure is related to methods of killing a tumor cell, including contacting a tumor cell with an effective amount of a composition including the antibody or antigen-binding fragment thereof, or the antibody-drug conjugate described herein.
As used herein, by an “effective amount” is meant an amount or dosage sufficient to effect beneficial or desired results including halting, slowing, retarding, or inhibiting progression of a disease, e.g., a cancer. An effective amount will vary depending upon, e.g., an age and a body weight of a subject to which the antibody, antigen binding fragment, antibody-encoding polynucleotide, vector comprising the polynucleotide, and/or compositions thereof is to be administered, a severity of symptoms and a route of administration, and thus administration can be determined on an individual basis.
An effective amount can be administered in one or more administrations. By way of example, an effective amount of an antibody or an antigen binding fragment thereof is an amount sufficient to ameliorate, stop, stabilize, reverse, inhibit, slow and/or delay progression of a disease in a patient. As is understood in the art, an effective amount of an antibody or antigen binding fragment may vary, depending on, inter alia, patient history as well as other factors such as the type (and/or dosage) of antibody used.
Effective amounts and schedules for administering the antibodies, antibody-encoding polynucleotides, and/or compositions disclosed herein may be determined empirically, and
making such determinations is within the skill in the art. Those skilled in the art will understand that the dosage that must be administered will vary depending on, for example, the mammal that will receive the antibodies, antibody-encoding polynucleotides, and/or compositions disclosed herein, the route of administration, the particular type of antibodies, antibody-encoding polynucleotides, antigen binding fragments, and/or compositions disclosed herein used and other drugs being administered to the mammal. Guidance in selecting appropriate doses for antibody or antigen binding fragment can be found in the literature on therapeutic uses of antibodies and antigen binding fragments, e.g., Handbook of Monoclonal Antibodies, Ferrone et al., eds., Noges Publications, Park Ridge, N. J., 1985, ch. 22 and pp. 303-357; Smith et al., Antibodies in Human Diagnosis and Therapy, Haber et al., eds., Raven Press, New York, 1977, pp. 365-389.
A typical daily dosage of an effective amount of an antibody is 0.01 mg/kg to 100 mg/kg. In some embodiments, the dosage can be less than 100 mg/kg, 10 mg/kg, 9 mg/kg, 8 mg/kg, 7 mg/kg, 6 mg/kg, 5 mg/kg, 4 mg/kg, 3 mg/kg, 2 mg/kg, 1 mg/kg, 0.5 mg/kg, or 0.1 mg/kg. In some embodiments, the dosage can be greater than 10 mg/kg, 9 mg/kg, 8 mg/kg, 7 mg/kg, 6 mg/kg, 5 mg/kg, 4 mg/kg, 3 mg/kg, 2 mg/kg, 1 mg/kg, 0.5 mg/kg, 0.1 mg/kg, 0.05 mg/kg, or 0.01 mg/kg. In some embodiments, the dosage is about 10 mg/kg, 9 mg/kg, 8 mg/kg, 7 mg/kg, 6 mg/kg, 5 mg/kg, 4 mg/kg, 3 mg/kg, 2 mg/kg, 1 mg/kg, 0.9 mg/kg, 0.8 mg/kg, 0.7 mg/kg, 0.6 mg/kg, 0.5 mg/kg, 0.4 mg/kg, 0.3 mg/kg, 0.2 mg/kg, or 0.1 mg/kg.
In any of the methods described herein, the at least one antibody, antigen-binding fragment thereof, or pharmaceutical composition (e.g., any of the antibodies, antigen-binding fragments, or pharmaceutical compositions described herein) and, optionally, at least one additional therapeutic agent can be administered to the subject at least once a week (e.g., once a week, twice a week, three times a week, four times a week, once a day, twice a day, or three times a day) . In some embodiments, at least two different antibodies and/or antigen-binding fragments are administered in the same composition (e.g., a liquid composition) . In some embodiments, at least one antibody or antigen-binding fragment and at least one additional therapeutic agent are administered in the same composition (e.g., a liquid composition) . In some embodiments, the at least one antibody or antigen-binding fragment and the at least one additional therapeutic agent are administered in two different compositions (e.g., a liquid composition containing at least one antibody or antigen-binding fragment and a solid oral composition containing at least one additional therapeutic agent) . In some embodiments, the at least one additional therapeutic agent is administered as a pill, tablet, or capsule. In some embodiments, the at least one additional therapeutic agent is administered in a sustained-release oral formulation.
In some embodiments, the one or more additional therapeutic agents can be administered to the subject prior to, or after administering the at least one antibody, antigen-binding antibody fragment, or pharmaceutical composition (e.g., any of the antibodies, antigen-binding antibody fragments, or pharmaceutical compositions described herein) . In some embodiments, the one or more additional therapeutic agents and the at least one antibody, antigen-binding antibody fragment, or pharmaceutical composition (e.g., any of the antibodies, antigen-binding antibody fragments, or pharmaceutical compositions described herein) are administered to the subject such that there is an overlap in the bioactive period of the one or more additional therapeutic agents and the at least one antibody or antigen-binding fragment (e.g., any of the antibodies or antigen-binding fragments described herein) in the subject.
In some embodiments, the subject can be administered the at least one antibody, antigen-binding antibody fragment, or pharmaceutical composition (e.g., any of the antibodies, antigen-
binding antibody fragments, or pharmaceutical compositions described herein) over an extended period of time (e.g., over a period of at least 1 week, 2 weeks, 3 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 12 months, 1 year, 2 years, 3 years, 4 years, or 5 years) . A skilled medical professional may determine the length of the treatment period using any of the methods described herein for diagnosing or following the effectiveness of treatment (e.g., the observation of at least one symptom of the disease) . As described herein, a skilled medical professional can also change the identity and number (e.g., increase or decrease) of antibodies or antigen-binding antibody fragments (and/or one or more additional therapeutic agents) administered to the subject and can also adjust (e.g., increase or decrease) the dosage or frequency of administration of at least one antibody or antigen-binding antibody fragment (and/or one or more additional therapeutic agents) to the subject based on an assessment of the effectiveness of the treatment (e.g., using any of the methods described herein and known in the art) .
In some embodiments, one or more additional therapeutic agents can be administered to the subject. The additional therapeutic agent can comprise one or more inhibitors selected from the group consisting of an inhibitor of B-Raf, an EGFR inhibitor, an inhibitor of a MEK, an inhibitor of ERK, an inhibitor of K-Ras, an inhibitor of c-Met, an inhibitor of anaplastic lymphoma kinase (ALK) , an inhibitor of a phosphatidylinositol 3-kinase (PI3K) , an inhibitor of an Akt, an inhibitor of mTOR, a dual PI3K/mTOR inhibitor, an inhibitor of Bruton's tyrosine kinase (BTK) , and an inhibitor of Isocitrate dehydrogenase 1 (IDH1) and/or Isocitrate dehydrogenase 2 (IDH2) .
In some embodiments, the additional therapeutic agent can comprise one or more inhibitors selected from the group consisting of an inhibitor of HER3, an inhibitor of LSD1, an inhibitor of MDM2, an inhibitor of BCL2, an inhibitor of CHK1, an inhibitor of activated hedgehog signaling pathway, and an agent that selectively degrades the estrogen receptor.
In some embodiments, the additional therapeutic agent can comprise one or more therapeutic agents selected from the group consisting of Trabectedin, nab-paclitaxel, Trebananib, Pazopanib, Cediranib, Palbociclib, everolimus, fluoropyrimidine, IFL, regorafenib, Reolysin, Alimta, Zykadia, Sutent, temsirolimus, axitinib, everolimus, sorafenib, Votrient, Pazopanib, IMA-901, AGS-003, cabozantinib, Vinflunine, an Hsp90 inhibitor, Ad-GM-CSF, Temazolomide, IL-2, IFNa, vinblastine, Thalomid, dacarbazine, cyclophosphamide, lenalidomide, azacytidine, lenalidomide, bortezomid, amrubicine, carfilzomib, pralatrexate, and enzastaurin.
In some embodiments, the additional therapeutic agent can comprise one or more therapeutic agents selected from the group consisting of an adjuvant, a TLR agonist, tumor necrosis factor (TNF) alpha, IL-1, HMGB1, an IL-10 antagonist, an IL-4 antagonist, an IL-13 antagonist, an IL-17 antagonist, an HVEM antagonist, an ICOS agonist, a treatment targeting CX3CL1, a treatment targeting CXCL9, a treatment targeting CXCL10, a treatment targeting CCL5, an LFA-1 agonist, an ICAM1 agonist, and a Selectin agonist.
In some embodiments, carboplatin, nab-paclitaxel, paclitaxel, cisplatin, pemetrexed, gemcitabine, FOLFOX, or FOLFIRI are administered to the subject.
In some embodiments, the additional therapeutic agent is an anti-PD-1 antibody, an anti-PD-L1 antibody, an anti-LAG-3 antibody, an anti-TIGIT antibody, an anti-4-1BB antibody, an anti-CTLA-4 antibody, an anti-CD40 antibody, an anti-OX40 antibody, or an anti-GITR antibody.
Pharmaceutical Compositions and Routes of Administration
Also provided herein are pharmaceutical compositions that contain at least one (e.g., one, two, three, or four) of the antibodies or antigen-binding fragments described herein. Two or more (e.g., two, three, or four) of any of the antibodies or antigen-binding fragments described herein can be present in a pharmaceutical composition in any combination. The pharmaceutical compositions may be formulated in any manner known in the art.
Pharmaceutical compositions are formulated to be compatible with their intended route of administration (e.g., intravenous, intraarterial, intramuscular, intradermal, subcutaneous, or intraperitoneal) . The compositions can include a sterile diluent (e.g., sterile water or saline) , a fixed oil, polyethylene glycol, glycerine, propylene glycol or other synthetic solvents, antibacterial or antifungal agents, such as benzyl alcohol or methyl parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like, antioxidants, such as ascorbic acid or sodium bisulfite, chelating agents, such as ethylenediaminetetraacetic acid, buffers, such as acetates, citrates, or phosphates, and isotonic agents, such as sugars (e.g., dextrose) , polyalcohols (e.g., mannitol or sorbitol) , or salts (e.g., sodium chloride) , or any combination thereof. Liposomal suspensions can also be used as pharmaceutically acceptable carriers (see, e.g., U.S. Patent No. 4,522,811) . Preparations of the compositions can be formulated and enclosed in ampules, disposable syringes, or multiple dose vials. Where required (as in, for example, injectable formulations) , proper fluidity can be maintained by, for example, the use of a coating, such as lecithin, or a surfactant. Absorption of the antibody or antigen-binding fragment thereof can be prolonged by including an agent that delays absorption (e.g., aluminum monostearate and gelatin) . Alternatively, controlled release can be achieved by implants and microencapsulated delivery systems, which can include biodegradable, biocompatible polymers (e.g., ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid; Alza Corporation and Nova Pharmaceutical, Inc. ) .
Compositions containing one or more of any of the antibodies or antigen-binding fragments described herein can be formulated for parenteral (e.g., intravenous, intraarterial, intramuscular, intradermal, subcutaneous, or intraperitoneal) administration in dosage unit form (i.e., physically discrete units containing a predetermined quantity of active compound for ease of administration and uniformity of dosage) .
Toxicity and therapeutic efficacy of compositions can be determined by standard pharmaceutical procedures in cell cultures or experimental animals (e.g., monkeys) . One can, for example, determine the LD50 (the dose lethal to 50%of the population) and the ED50 (the dose therapeutically effective in 50%of the population) : the therapeutic index being the ratio of LD50: ED50. Agents that exhibit high therapeutic indices are preferred. Where an agent exhibits an undesirable side effect, care should be taken to minimize potential damage (i.e., reduce unwanted side effects) . Toxicity and therapeutic efficacy can be determined by other standard pharmaceutical procedures.
Data obtained from cell culture assays and animal studies can be used in formulating an appropriate dosage of any given agent for use in a subject (e.g., a human) . A therapeutically effective amount of the one or more (e.g., one, two, three, or four) antibodies or antigen-binding fragments thereof (e.g., any of the antibodies or antibody fragments described herein) will be an amount that treats the disease in a subject in a subject, or a subject identified as being at risk of developing the disease, decreases the severity, frequency, and/or duration of one or more symptoms of a disease in a subject (e.g., a human) . The effectiveness and dosing of any of the antibodies or antigen-binding fragments described herein can be determined by a health care professional or veterinary professional using methods known in the art, as well as by the
observation of one or more symptoms of disease in a subject (e.g., a human) . Certain factors may influence the dosage and timing required to effectively treat a subject (e.g., the severity of the disease or disorder, previous treatments, the general health and/or age of the subject, and the presence of other diseases) .
Exemplary doses include milligram or microgram amounts of any of the antibodies or antigen-binding fragments described herein per kilogram of the subject’s weight (e.g., about 1 μg/kg to about 500 mg/kg; about 100 μg/kg to about 500 mg/kg; about 100 μg/kg to about 50 mg/kg; about 10 μg/kg to about 5 mg/kg; about 10 μg/kg to about 0.5 mg/kg; or about 1 μg/kg to about 50 μg/kg) . While these doses cover a broad range, one of ordinary skill in the art will understand that therapeutic agents, including antibodies and antigen-binding fragments thereof, vary in their potency, and effective amounts can be determined by methods known in the art. Typically, relatively low doses are administered at first, and the attending health care professional or veterinary professional (in the case of therapeutic application) or a researcher (when still working at the development stage) can subsequently and gradually increase the dose until an appropriate response is obtained. In addition, it is understood that the specific dose level for any particular subject will depend upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, gender, and diet of the subject, the time of administration, the route of administration, the rate of excretion, and the half-life of the antibody or antibody fragment in vivo.
The pharmaceutical compositions can be included in a container, pack, or dispenser together with instructions for administration. The disclosure also provides methods of manufacturing the antibodies or antigen binding fragments thereof for various uses as described herein.
EXAMPLES
The invention is further described in the following examples, which do not limit the scope of the invention described in the claims.
EXAMPLE 1. Generating human anti-TFR1 antibodies
RenNanoTM mice (Biocytogen, complete human heavy chain variable domain substitution in situ, combined with a modified constant region. The mice are described e.g., in PCT/CN2022/119188, which is incorporated herein by reference in its entirety) were immunized with His-tagged human TFR1 (transferrin receptor 1) protein (hTFR1-His, ACROBiosystems, Cat#: CD1-H5243) to obtain anti-TFR1 antibodies. Before immunization, retro-orbital blood was collected as a negative control. Freund’s complete adjuvant (CFA) was used for the first immunization and Freund’s incomplete adjuvant (IFA) was used for the second and third immunizations. A total of three immunizations (bi-weekly) were performed. One week after the third immunization, retro-orbital blood was collected, and the antibody titer of serum was detected by FACS.
Procedures to enhance immunization were also performed at least fourteen days after the previous immunizations. TFR1 protein was injected by intraperitoneal injection, and the CHO-Scells expressing human TFR1 antigen was injected through the tail vein.
Antigen-specific immune cells were isolated from the immunized mice to further obtain anti-TFR1 antibodies or to obtain the heavy chain variable region sequences of the anti-TFR1 antibodies. For example, single cell technology (e.g., usingOptofluidic System, Berkeley Lights Inc. ) was used to screen and find plasma cells that secrete antigen-specific
monoclonal antibodies. Reverse transcription and PCR sequencing were used to obtain antibody variable region sequences. The obtained variable region sequences were used for antibody expression to verify the binding activity to TFR1 using FACS. Because the lack of the CH1 domain, the heavy chain variable region (VH) of the obtained antibodies is also referred to as a heavy chain single variable domain (VHH) .
Specifically, the obtained VHH sequences were respectively connected to a human IgG1 constant region (e.g., the hinge region, CH2 domain and CH3 domain) . Exemplary antibodies obtained by this method included: 23B8, 24A1, 24C9 and 24G5. The heavy chain CDR1-3 sequences are shown in FIG. 1 and FIG. 2. The VHH region sequences of 23B8, 24A1, 24C9 and 24G5 are shown in FIG. 3.
The constant region of the antibodies can be further engineered to replace the Asparagine at position 297 with Alanine (N297A) . For example, when N297A mutation is introduced into the constant region of 24G5, the resulting antibody is named as 24G5-N.
Further, one-armed antibodies 23B8-mono, 24A1-mono, 24C9-mono and 24G5-mono were constructed, these antibodies have an anti-TFR1 arm comprising a VHH region, and a heavy chain fragment comprising CH2 and CH3 domains of IgG1 with N297A mutation.
EXAMPLE 2. Cross-species binding of anti-TFR1 antibodies
CHO-S-hTFR1 cells or CHO-S-fasTFR1 cells were transferred to a 96-well plate at a density of 105 cells/well respectively. Serially diluted sample anti-TFR1 antibodies were added to the 96-well plate, and incubated at 4℃ for 30 minutes. PBS was used as a negative control (NC) . Then, the cells were incubated with the secondary antibody anti-hIgG-Fc-Alex FlourTM 647 (Jackson ImmunoResearch Laboratories, Cat#: 109-606-170) at 4℃ in the dark for 15 minutes before flow cytometry analysis.
CHO-S-hTFR1 cells or CHO-S-fasTFR1 cells were obtained by transfecting CHO-Scells with vectors expressing human TFR1 (hTFR1, SEQ ID NO: 29) or Macaca fascicularis (crab-eating macaque) TFR1 amino acid sequence (fasTFR1, SEQ ID NO: 30) , respectively. The test results are shown in the table below.
JR141, a humanized IgG1 antibody targeting human TFR1 conjugated to human iduronate-2-sulfatase, was first approved in March 2021 in Japan for the intravenous treatment of mucopolysaccharidosis type II. The VH and VL sequences of JR141 are set forth in SEQ ID NO: 31 and SEQ ID NO: 32, respectively. For positive control (JR141-N) , the VH and VL of JR141 were connected to a human IgG1 constant region with N297A mutation.
Table 1
EXAMPLE 3. Binding affinity of anti-TFR1 antibodies
The binding affinity of the anti-TFR1 antibodies to His-tagged TFR1 protein of human (hTFR1-His, ACROBiosystems, Cat#: CD1-H5243) or monkey (fasTFR1-His, ACROBiosystems, Cat#: TFR-C524a) were verified using surface plasmon resonance (SPR) on BiacoreTM (Biacore, Inc., Piscataway N. J. ) 8K biosensor equipped with pre-immobilized Protein A sensor chips.
Purified anti-TFR1 antibodies was captured on the Protein A chip (Series S Sensor Chip Protein A) for the detection. Purified anti-TFR1 antibodies (1 μg/mL) was loaded at 10 μL/min to bind to hTFR1-His and fasTFR1-His (200 nM) . The flow rate was 30 μL/min. The binding and dissociation time were set to 180 seconds and 600 seconds, respectively. The chip was regenerated after the last injection of each titration with a glycine solution (pH 2.0) at 30 μL/min for 30 seconds.
Kinetic association rates (kon) and dissociation rates (koff) were obtained simultaneously by fitting the data globally to a 1: 1 Langmuir binding model (Karlsson, R. Roos, H. Fagerstam, L. Petersson, B., 1994. Methods Enzymology 6.99-110) using BiacoreTM8K Evaluation Software 3.0. Affinities were deduced from the quotient of the kinetic rate constants (KD=koff/kon) .
As a person of ordinary skill in the art would understand, the same method with appropriate adjustments for parameters (e.g., antibody concentration) was performed for each tested antibody. The results for the tested antibodies are summarized in the table below. The results show that all four anti-TFR1 antibodies can bind to human and monkey TFR1 with high affinity.
Table 2
EXAMPLE 4. Epitope analysis of anti-TFR1 antibodies
Relative positions of target protein epitope between a pair of purified anti-TFR1 antibodies were analyzed by Biolayer Interferometry (BLI) using ForteBio Octet system at 30 ℃. 1× HBS-EP+ buffer (10 mM 4- (2-hydroxyethyl) -1-piperazineethanesulfonic acid (HEPES) , 150 mM NaCl, 3 mM ethylenediaminetetraacetic acid (EDTA) and 0.05%P20, pH7.4) diluted from HBS-EP+ buffer (10×) was used as the running buffer throughout the experiment. About 10 μg/mL of hTFR1-His protein was captured by HIS1K (Anti-Penta-HIS) for 200 seconds, and 200 nM of antibody (analyte 1) was injected at a flow rate of 30 μL/min to bind the ligand. Another antibody (analyte 2) was injected under the same conditions to determine whether the binding of different antibodies interfered with each other. The binding time was 300 seconds for each antibody.
The binding value of each antibody was obtained using Data Analysis HT 12.0. To quantify the interference of one antibody binding to another, a binding ratio was calculated to
compare each pair of antibodies. The binding ratio is defined as the binding value of the second antibody (analyte 2) , divided by the binding value of the first antibody (analyte 1) . The binding ratio of each antibody pair is summarized in the matrix table below. Specifically, the binding ratio was between 0.0 to 0.5, if analyte 1 exhibited a blocking effect to analyte 2; the binding ratio was between 0.5-1.1, if analyte 1 did not exhibit a blocking effect to analyte 2. In general, antibody pairs that interfere with each other have the same or overlapping epitopes.
The epitope binding assay results indicate that 24A1 and 24G5 can recognize the same epitope, and 23B8, 24C9 and JR141-N can recognize different epitopes.
Table 3
EXAMPLE 5. Internalization of anti-TFR1 antibodies
Anti-TFR1 antibodies together with the pHAb-Goat anti-human IgG secondary antibody were added to human cortical microvascular endothelial cells (hCMEC/D3 cells) , and incubated for 3 hours. After incubation, the cells were centrifuged and washed with FACS buffer. Mean fluorescence intensity (MFI) was measured using a flow cytometer. Endocytosis rates of antibodies were calculated. Human IgG1 protein (CrownBio, Cat#: C0001) was used as an isotype control (ISO) . The results are shown in the following table, indicating that all four antibodies exhibited good endocytosis activity in human cortical microvascular endothelial cells.
Table 4
EXAMPLE 6. Developability analysis of anti-TFR1 antibodies
Developability of anti-TFR1 antibodies 23B8, 24A1, 24C9 and 24G5 was evaluated. The antibodies were diluted to 1 mg/mL with water. Specifically, the following tests were performed: (1) observing the solution appearance and presence of visible non-soluble objects; (2) detecting the purity changes of antibodies by Size-Exclusion Ultra Performance Liquid Chromatography (SEC-UPLC) (indicated as the percentage of the main peak area to the sum of all peak areas (Purity, %) ) ; (3) detecting changes in the apparent hydrophobicity of the antibodies using the Hydrophobic Interaction Chromatography-High Performance Liquid Chromatography (HIC-HPLC) method (indicated as the retention time of the main peak (HIC, min) ; (4) detecting charge variants in the antibodies by Capillary Isoelectric Focusing (cIEF) (indicated as the percentages of the main component, acidic component, and alkaline component) ; and (5) detecting the
thermal stability of antibodies via the UNcle system (indicated as the melting temperature (Tm) and aggregation temperature (Tagg) ) .
In the SEC-UPLC experiments, the Agilent 1290 chromatograph system (connected with XBridgeTM Protein BEH SEC column (Waters Corporation) ) was used. The antibody samples were diluted to 1 mg/mL with purified water. The following parameters were used: mobile phase: 25 mM phosphate buffer (PB) (pH 6.8) + 0.3 M NaCl; flow rate: 1.8 mL/min; column temperature: 25 ℃; detection wavelength: 280 nm; injection volume: 10 μL; sample tray temperature: 6 ℃; and running time: 7 minutes.
In the HIC-HPLC experiments, the Agilent 1260 chromatograph system (connected with ProPacTM HIC-10 column (4.6 × 100 mm, Thermo Scientific) ) was used, and samples were diluted using mobile phase A to 0.5 mg/mL. The following parameters were used: mobile phase A: 0.9 M ammonium sulfate, 0.1 M PB, 10%acetonitrile pH 6.5; mobile phase B: 0.1 M PB, 10%acetonitrile pH 6.5; flow rate: 0.8 mL/min; gradient: 0 min 100%A, 2 min 100%A, 32 min 100%B, 34 min 100%B, 35 min 100%A, and 45 min 100%A; column temperature: 30 ℃; detection wavelength: 280 nm; injection volume: 10 μg; sample tray temperature: about 6 ℃; and running time: 45 minutes.
In the cIEF experiments, the Maurice cIEF Method Development Kit (Protein Simple, Cat#: PS-MDK01-C) was used for sample preparation. Specifically, 40 μg protein sample was mixed with the following reagents in the kit: 1 μL Maurice cIEF pI Marker-4.05, 1 μL Maurice cIEF pI Marker-9.99, 35 μL 1%Methyl Cellulose Solution, 2 μL Maurice cIEF 500 mM Arginine, 4 μL Ampholytes (Pharmalyte pH ranges 3-10) , and water (added to make a final volume of 100 μL) . On the Maurice analyzer (Protein Simple, Santa Clara, CA) , Maurice cIEF Cartridges (PS-MC02-C) were used to generate imaging capillary isoelectric focusing spectra. The sample was focused for a total of 10 minutes. The analysis software installed on the instrument was used to analyze the absorbance of the 280 nm-focused protein.
In the thermal stability experiments, antibody solutions at 60 mg/mL were heated from 25℃ to 95 ℃ using 1 ℃ increments, with an equilibration time of 1 minute before each measurement.
Furthermore, the antibodies were diluted to 1 mg/mL using PBS buffer, and the following tests were performed: (1) detecting the specificity of the antibodies using the Cross-Interaction Chromatography (CIC) method (indicated as the retention time (CIC, min) ) ; (2) detecting the colloidal stabilities of the antibodies by the stand-up monolayer chromatography (SMAC) method (indicated as the retention time (SMAC, %/min) ) .
In the CIC assay, a CIC column was prepared by coupling human polyclonal IgGs (Sigma, Cat #: I4506) onto a HiTrap NHS-activated resin (GE Healthcare, Cat #: 17-0716-01) followed by passivation with ethanolamine according to published procedures. The column was then connected to Agilent 1260 chromatograph system and run at 0.1 mL/min using 1× PBS as the mobile phase until a flat baseline was reached. 10 μg of antibodies at 1 mg/mL in PBS were then injected. Peak retention times on the column were monitored at 280 nm; running time: 50 minutes.
In the SMAC assay, Zenix column (4.6 mm × 30 cm, Sepax, Cat #: 213300-4630) was connected to the column compartment, and place the appropriate line in the mobile phase. Equilibrate column for 60 min at 0.350 mL/min flow rate with mobile phase buffer. Load antibodies into the injection sequence. Mobile Phase A: 150 mM Sodium Phosphate pH 7.0; flow rate: 0.35 mL/min; running time: 25 min; column temperature: 30℃; detection wavelength: 280 nm, 220 nm.
Detailed results are summarized in the table below.
Table 5
For the Tagg of 24C9, a slight fluctuation was observed at 33.63 ℃ from the Tagg curve (data not shown) , while massive aggregation occured at 50-60 ℃. To confirm the thermal stability, SEC-UPLC of 24C9 was tested after treatment of 40 ℃ for 14 days. After heating, the purity of 24C9 stayed above 97%. These results indicated that all the four antibodies have good developabilities.
EXAMPLE 7. Pharmacokinetics (PK) analysis
A humanized TFR1 mouse model (hTFR1 mice) was engineered to express a chimeric TFR1 protein (SEQ ID NO: 33) in which the extracellular region of mouse TFR1 protein was replaced with the corresponding human TFR1 extracellular region. A detailed description regarding the humanized TFR1 mouse model can be found in PCT Application No. PCT/CN2022/105924, which is incorporated herein by reference in its entirety.
The concentrations of the anti-TFR1 antibodies were determined in hTFR1 mice. Specifically, the mice were placed into different groups (8 mice per group) , and administered with an approximately equal molar dosage of JR141-N (G2) , 23B8-N (G3) , 24A1-N (G4) , 24G5-N (G5) , or 24C9-N (G6) , by intravenous (i. v. ) injection. The control group (G1) mice were administered with human IgG1 (hIgG1) . Details of the administration scheme are shown in the table below.
Table 6
Blood and brain samples were collected 0.5, 6, 24, and 72 hours after the administration. Two mice were sampled at each time point, and the mice were anesthetized after retro-orbital blood was collected. To avoid interference from the residual blood in the brain, the mice were perfused by saline for 10 minutes at room temperature. Specifically, saline was perfused via systemic circulation from the left ventricle to the right ventricle. Brain samples were excised and divided into two hemibrains by the sagittal plane. The left hemibrain was subjected to quantification of the injected antibody, while the right hemibrain was fixed by formalin and embedded with paraffin for serial sections. The brain samples were cut into pieces and homogenized with DPBS (Dulbecco's phosphate-buffered saline) containing 1× mixed protease inhibitors. The brain homogenate was aliquoted for protein extraction followed by antibody quantification by electrochemiluminescence. For the rest homogenate, capillaries were depleted
by gradient density centrifugation at 5400 g for 15 minutes, using 15%dextran. After centrifugation, the fraction at the top of the centrifuge tube was saved as parenchyma and subjected to protein extraction and antibody quantification as well. FIGS. 4A-4D show the antibody concentration in total brain protein (FIG. 4A) , the ratio of antibody concentration in brain total protein to serum antibody concentration (FIG. 4B) , the antibody concentration in brain parenchyma (FIG. 4C) , and the ratio of antibody concentration in brain parenchyma to serum antibody concentration at each time point (FIG. 4D) . These results demonstrated that 24G5-N (group G5) was the most abundant either in parenchyma or in the whole brain.
In a similar experiment, hTFR1 mice were placed into five groups (3 mice per group) and administered with 18.4 mg/kg JR141-N (G2) , 10 mg/kg 23B8-N (G3) , 10 mg/kg 24A1-N (G4) , or 10 mg/kg 24G5-N (G5) by intravenous injection (1 administration in total) . The control group (G1) mice were administered with hIgG1 (G1) . 24 hours after administration, brain samples were collected to determine the concentrations of the anti-TFR1 antibodies. FIGS. 5A-5B show the antibody concentration test results in brain parenchyma and brain total protein, respectively. All tested antibodies showed a higher concentration in brain than hIgG1 (G1) , indicating that compared with the positive control JR141-N (G2) , 23B8-N (G3) and 24G5-N (G5) can better pass across the blood-brain barrier and enter the brain parenchyma.
In another similar experiment, hTFR1 mice were placed into seven groups (6 mice per group) and administered with JR141-N (G2-G4) or 24G5-N (G5-G7) ) by intravenous (i. v. ) injection. The control group (G1) mice were administered with hIgG1. Details of the administration scheme are shown in the table below.
Table 7
6 hours and 24 hours after the administration, blood and brain samples were collected using the methods described above. Three mice were sampled at each time point. Treatment of the tissues and quantification of antibodies were also performed as described above. The measurement results of the concentration of humanized anti-TFR1 antibodies in the brain parenchyma are shown in FIG. 6. The results show that the antibody concentration accumulated in the brain parenchyma of 24G5-N was significantly higher than that of hIgG1 under each dosage condition. Furthermore, the concentration of both JR141-N and 24G5-N exhibited a dose-dependent trend in brain parenchyma.
To detect the distribution of the humanized anti-TFR1 antibody 24G5-N in mouse brain, immunofluorescence assays were performed by staining hIgG, hTFR1, and mCD31, respectively, on right hemibrain sections of the mice used in the experiment above. The results showed that, mCD31 was well-labeled in the microvessels. hTFR1 was also detected on microvessels, which was co-localized with mCD31. Besides, hTFR1 expression was also detected on several neurons in parenchyma. In particular, the anti-TFR1 antibody 24G5-N was stained by a secondary anti-
IgG antibody conjugated with488. Similar to hTFR1, 24G5-N was detected in microvessels and parenchyma, and its signal overlapped with the hTFR1 signal. Thus, either for the quantification of 24G5-N in the whole brain or parenchyma, or the visual evidence of immunofluorescence of 24G5-N in the parenchyma, the results indicate that anti-TFR1 antibody 24G5-N can pass across the blood-brain barrier (BBB) efficiently.
EXAMPLE 8. Blocking assay
Blockade of TFR1 binding to TF (transferrin) by anti-TFR1 antibodies 23B8, 24A1, 24C9 and 24G5 were tested by Biolayer Interferometry (BLI) using ForteBiosystem at 30 ℃. Specifically, 1× HBS-EP+ buffer (10 mM 4- (2-hydroxyethyl) -1-piperazineethanesulfonic acid (HEPES) , 150 mM NaCl, 3 mM EDTA and 0.05%Surfactant P20, pH 7.4) diluted from HBS-EP+ buffer (10×) was used as the running buffer throughout the experiment. About 10 μg/mL of antibodies were captured by AHC (Anti-Human IgG Fc Capture) for 200 seconds, and 800 nM of hTFR1-His (ACROBiosystems, Cat#: CD1-H5243) and hTF-His (human transferrin protein, Kactus Biosystems, Cat#: TFN-HM101) were injected to bind the ligand. The binding time was 300 seconds for each antibody. The binding value of each antibody was obtained using Data Analysis HT 12.0. The results showed that these four antibodies did not block the binding of TFR1 to TF. Therefore, such non-blocking antibodies are not likely to interfere with TFR1-TF interactions in normal cells.
EXAMPLE 9. Antibody Drug Conjugates (ADC)
Each purified antibody (23B8-N, 24A1-N, 24C9-N, 24G5-N, 23B8-mono, 24A1-mono, 24C9-mono and 24G5-mono) was coupled with Dxd (Deruxtecan) through GGFG linker.
For the names of antibody drug conjugates, “ADC” is added directly after the antibody name. For example, if 24G5-N is coupled to GGFG-Dxd, it is named as 24G5-ADC. For another examples, if 24G5-mono is coupled to GGFG-Dxd, it is named as 24G5-mono-ADC. HIC-HPLC (Reversed Phase High Performance Liquid Chromatography) was used to detect the coupling of antibodies with drug molecules. The HIC-HPLC detection results showed that the drug-to-antibody ratio (DAR) of the ADCs was about 3.
hTFR1 mice were placed into different groups (6 mice per group) and administered with 10 mg/kg bivalent anti-TFR1 ADCs (24G5-ADC, 23B8-ADC, 24A1-ADC, 24C9-ADC) or the same molar dose 8.34 mg/kg one-armed monovalent anti-TFR1 ADCs (23B8-mono-ADC, 24A1-mono-ADC, 24C9-mono-ADC, or 24G5-mono-ADC) by intravenous (i. v. ) injection (1 administration in total) . 0.5 hour, 18 hours and 72 hours after the administration, blood and brain samples were collected using the methods described in Example 7. Treatment of the tissues and quantification of antibodies and ADCs were also performed as described above The results indicated that all the tested ADCs are able to bring Dxd across the BBB efficiently. Exemplary results are shown in FIG. 7, 24G5-ADC and 24G5-mono-ADC bring Dxd across the BBB, of which the monovalent 24G5-mono-ADC has a better penetration effect than the bivalent format 24G5-ADC.
OTHER EMBODIMENTS
It is to be understood that while the invention has been described in conjunction with the detailed description thereof, the foregoing description is intended to illustrate and not limit the scope of the invention, which is defined by the scope of the appended claims. Other aspects, advantages, and modifications are within the scope of the following claims.
Claims (39)
- An antibody or antigen-binding fragment thereof that binds to transferrin receptor 1 (TFR1) , comprising:a heavy chain single variable domain (VHH) comprising complementarity determining regions (CDRs) 1, 2, and 3, wherein the VHH CDR1 region comprises an amino acid sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identical to a selected VHH CDR1 amino acid sequence, the VHH CDR2 region comprises an amino acid sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identical to a selected VHH CDR2 amino acid sequence, and the VHH CDR3 region comprises an amino acid sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identical to a selected VHH CDR3 amino acid sequence;wherein the selected VHH CDRs 1, 2, and 3 amino acid sequences are one of the following:(1) the selected VHH CDRs 1, 2, 3 amino acid sequences are set forth in SEQ ID NOs: 1, 2, and 3, respectively;(2) the selected VHH CDRs 1, 2, 3 amino acid sequences are set forth in SEQ ID NOs: 4, 5, and 6, respectively;(3) the selected VHH CDRs 1, 2, 3 amino acid sequences are set forth in SEQ ID NOs: 7, 8, and 9, respectively;(4) the selected VHH CDRs 1, 2, 3 amino acid sequences are set forth in SEQ ID NOs: 10, 11, and 12, respectively;(5) the selected VHH CDRs 1, 2, 3 amino acid sequences are set forth in SEQ ID NOs: 13, 14, and 15, respectively;(6) the selected VHH CDRs 1, 2, 3 amino acid sequences are set forth in SEQ ID NOs: 16, 17, and 18, respectively;(7) the selected VHH CDRs 1, 2, 3 amino acid sequences are set forth in SEQ ID NOs: 19, 20, and 21, respectively; and(8) the selected VHH CDRs 1, 2, 3 amino acid sequences are set forth in SEQ ID NOs: 22, 23, and 24, respectively.
- The antibody or antigen-binding fragment thereof of claim 1, wherein the VHH comprises CDRs 1, 2, 3 with the amino acid sequences set forth in SEQ ID NOs: 1, 2, and 3, respectively.
- The antibody or antigen-binding fragment thereof of claim 1, wherein the VHH comprises CDRs 1, 2, 3 with the amino acid sequences set forth in SEQ ID NOs: 4, 5, and 6, respectively.
- The antibody or antigen-binding fragment thereof of claim 1, wherein the VHH comprises CDRs 1, 2, 3 with the amino acid sequences set forth in SEQ ID NOs: 7, 8, and 9, respectively.
- The antibody or antigen-binding fragment thereof of claim 1, wherein the VHH comprises CDRs 1, 2, 3 with the amino acid sequences set forth in SEQ ID NOs: 10, 11, and 12, respectively.
- An antibody or antigen-binding fragment thereof that binds to TFR1 comprising a heavy chain single variable region (VHH) comprising an amino acid sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identical to a selected VHH sequence, wherein the selected VHH sequence is selected from the group consisting of SEQ ID NOs: 25, 26, 27, and 28.
- The antibody or antigen-binding fragment thereof of claim 6, wherein the VHH comprises the sequence of SEQ ID NO: 25.
- The antibody or antigen-binding fragment thereof of claim 6, wherein the VHH comprises the sequence of SEQ ID NO: 26.
- The antibody or antigen-binding fragment thereof of claim 6, wherein the VHH comprises the sequence of SEQ ID NO: 27.
- The antibody or antigen-binding fragment thereof of claim 6, wherein the VHH comprises the sequence of SEQ ID NO: 28.
- The antibody or antigen-binding fragment thereof of any one of claims 1-10, wherein the antibody or antigen-binding fragment specifically binds to a human TFR1, a monkey TFR1, a mouse TFR1, or a chimeric TFR1.
- The antibody or antigen-binding fragment thereof of any one of claims 1-11, wherein the antibody or antigen-binding fragment is a human or humanized antibody or antigen-binding fragment thereof.
- The antibody or antigen-binding fragment thereof of any one of claims 1-12, wherein the antibody or antigen-binding fragment is a multi-specific antibody (e.g., a bispecific antibody) .
- An antibody or antigen-binding fragment thereof comprising the VHH CDRs 1, 2, 3, of the antibody or antigen-binding fragment thereof of any one of claims 1-13.
- The antibody or antigen-binding fragment thereof of any one of claims 1-14, wherein the antibody or antigen-binding fragment comprises a human IgG Fc (e.g., a human IgG1 Fc) .
- The antibody or antigen-binding fragment thereof of claim 15, wherein the human IgG Fc comprises a non-asparagine residue (e.g., alanine) at position 297 according to EU numbering.
- The antibody or antigen-binding fragment thereof of any one of claims 1-16, wherein the antibody or antigen-binding fragment comprises two or more heavy chain single variable domains.
- A nucleic acid comprising a polynucleotide encoding the antibody or antigen-binding fragment thereof any one of claims 1-17.
- The nucleic acid of claim 18, wherein the nucleic acid is cDNA.
- A vector comprising one or more of the nucleic acids of claim 18 or 19.
- A cell comprising the vector of claim 20.
- The cell of claim 21, wherein the cell is a CHO cell.
- A cell comprising one or more of the nucleic acids of claims 18 or 19.
- A method of producing an antibody or an antigen-binding fragment thereof, the method comprising(a) culturing the cell of any one of claims 21-23 under conditions sufficient for the cell to produce the antibody or the antigen-binding fragment thereof; and(b) collecting the antibody or the antigen-binding fragment thereof produced by the cell.
- An antibody-drug conjugate comprising the antibody or antigen-binding fragment thereof of any one of claims 1-17 covalently bound to a therapeutic agent.
- The antibody drug conjugate of claim 25, wherein the therapeutic agent is a cytotoxic or cytostatic agent.
- A method of treating a subject having a brain disease (e.g., a brain cancer) , the method comprising administering a therapeutically effective amount of a composition comprising the antibody or antigen-binding fragment thereof of any one of claims 1-17, or the antibody-drug conjugate of claim 25 or 26, to the subject.
- The method of claim 27, wherein the antibody or antigen-binding fragment thereof, or the antibody-drug conjugate can pass across the blood-brain barrier (BBB) of the subject.
- A method of treating a subject having a cancer, the method comprising administering a therapeutically effective amount of a composition comprising the antibody or antigen-binding fragment thereof of any one of claims 1-17, or the antibody-drug conjugate of claim 25 or 26, to the subject.
- The method of claim 29, wherein the cancer is brain cancer, lung cancer, gastric cancer, colorectal cancer, liver cancer, ovarian cancer, prostate cancer, leukemia, or breast cancer.
- A method of identifying a subject as having a brain disease (e.g., a brain cancer) , the method comprisingdetecting a sample collected from the subject as having the brain disease by the antibody or antigen-binding fragment thereof of any one of claims 1-17,thereby identifying the subject as having the brain disease.
- The method of claim 31, wherein the sample is a brain parenchyma sample from the subject.
- The method of any one of claims 27-32, wherein the subject is a human subject.
- A method of delivering an agent to cross blood brain barrier, the method comprisingadministering the agent covalently linked to the antibody or antigen-binding fragment thereof of any one of claims 1-17 to the subject.
- The method of claim 34, wherein the agent is an antibody or an antibody drug conjugate.
- The method of claim 34 or 35, wherein the agent is anti-amyloid antibody.
- A pharmaceutical composition comprising the antibody or antigen-binding fragment thereof of any one of claims 1-17, and a pharmaceutically acceptable carrier.
- A pharmaceutical composition comprising the antibody drug conjugate of claim 25 or 26, and a pharmaceutically acceptable carrier.
- An antibody or antigen-binding fragment thereof that cross-competes with the antibody or antigen-binding fragment thereof of any one of claims 1-17.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN2022136246 | 2022-12-02 | ||
CNPCT/CN2022/136246 | 2022-12-02 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2024114687A1 true WO2024114687A1 (en) | 2024-06-06 |
Family
ID=91323009
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/CN2023/135130 WO2024114687A1 (en) | 2022-12-02 | 2023-11-29 | Anti-tfr1 antibodies and uses thereof |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2024114687A1 (en) |
Citations (6)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20060039908A1 (en) * | 2004-06-07 | 2006-02-23 | Mather Jennie P | Transferrin receptor antibodies |
US20060130158A1 (en) * | 2002-08-30 | 2006-06-15 | Turner Andrew J | Modified transferrin fusion proteins comprising duplicate transferrin amino or carboxy terminal domains |
US20130171061A1 (en) * | 2011-12-29 | 2013-07-04 | Ming-Hua Yang | Anti-human transferrin receptor antibody and uses thereof |
JP2014093958A (en) * | 2012-11-08 | 2014-05-22 | Univ Of Miyazaki | Antibody capable of specifically recognizing transferrin receptor |
WO2017013230A1 (en) * | 2015-07-22 | 2017-01-26 | Inatherys | ANTI-TfR ANTIBODIES AND THEIR USE IN TREATING PROLIFERATIVE AND INFLAMMATORY DISORDERS |
WO2020144233A1 (en) * | 2019-01-09 | 2020-07-16 | Vect-Horus | Transferrin receptor-binding molecules, conjugates thereof and their uses |
-
2023
- 2023-11-29 WO PCT/CN2023/135130 patent/WO2024114687A1/en unknown
Patent Citations (7)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20060130158A1 (en) * | 2002-08-30 | 2006-06-15 | Turner Andrew J | Modified transferrin fusion proteins comprising duplicate transferrin amino or carboxy terminal domains |
US20060039908A1 (en) * | 2004-06-07 | 2006-02-23 | Mather Jennie P | Transferrin receptor antibodies |
US20130171061A1 (en) * | 2011-12-29 | 2013-07-04 | Ming-Hua Yang | Anti-human transferrin receptor antibody and uses thereof |
JP2014093958A (en) * | 2012-11-08 | 2014-05-22 | Univ Of Miyazaki | Antibody capable of specifically recognizing transferrin receptor |
WO2017013230A1 (en) * | 2015-07-22 | 2017-01-26 | Inatherys | ANTI-TfR ANTIBODIES AND THEIR USE IN TREATING PROLIFERATIVE AND INFLAMMATORY DISORDERS |
US20220119543A1 (en) * | 2015-07-22 | 2022-04-21 | Inatherys | ANTI-TfR ANTIBODIES AND THEIR USE IN TREATING PROLIFERATIVE AND INFLAMMATORY DISORDERS |
WO2020144233A1 (en) * | 2019-01-09 | 2020-07-16 | Vect-Horus | Transferrin receptor-binding molecules, conjugates thereof and their uses |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
US11155625B2 (en) | Anti-PD-1 antibodies and uses thereof | |
US20240002539A1 (en) | Multispecific antibodies and uses thereof | |
CN113227142B (en) | anti-PD-1 antibodies and uses thereof | |
CN112533950B (en) | anti-CD 3e antibodies and uses thereof | |
US20240109963A1 (en) | Multispecific antibodies and uses thereof | |
WO2021098851A1 (en) | Anti-ctla4/ox40 bispecific antibodies and uses thereof | |
KR20200088810A (en) | Anti-OX40 antibodies and uses thereof | |
US20230399408A1 (en) | Multispecific antibodies and uses thereof | |
CN112739718A (en) | anti-TNFRSF 9 antibodies and uses thereof | |
WO2023143343A1 (en) | Anti-her2/trop2 antibodies and uses thereof | |
WO2024114687A1 (en) | Anti-tfr1 antibodies and uses thereof | |
WO2023078450A1 (en) | Multispecific antibodies and uses thereof | |
WO2024175020A1 (en) | Anti-il2ra antibodies and uses thereof | |
WO2023241538A1 (en) | Anti-siglec15 antibodies and uses thereof | |
WO2024208244A1 (en) | Anti-igf1r antibodies and uses thereof | |
WO2024056044A1 (en) | Genetically modified non-human animals and methods for producing heavy-chain antibodies | |
WO2023217289A1 (en) | Multispecific antibodies and uses thereof | |
WO2023116813A1 (en) | Anti-tnfr2 antibodies and uses thereof | |
WO2024044732A2 (en) | Multispecific antibodies and uses thereof | |
TW202432594A (en) | Anti-ccr8 antibodies and uses thereof | |
WO2023196598A2 (en) | Anti-mica/b antibodies and uses thereof | |
TW202434646A (en) | Anti-il2ra antibodies and uses thereof | |
CN118541390A (en) | Anti-CTLA 4/OX40 bispecific antibodies and uses thereof |
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: 23896831 Country of ref document: EP Kind code of ref document: A1 |