EP4493586A1 - Breit neutralisierende antikörper gegen hepatitis-e-virus - Google Patents
Breit neutralisierende antikörper gegen hepatitis-e-virusInfo
- Publication number
- EP4493586A1 EP4493586A1 EP23710899.8A EP23710899A EP4493586A1 EP 4493586 A1 EP4493586 A1 EP 4493586A1 EP 23710899 A EP23710899 A EP 23710899A EP 4493586 A1 EP4493586 A1 EP 4493586A1
- Authority
- EP
- European Patent Office
- Prior art keywords
- sequence identity
- seq
- antibody
- construct
- hev
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Pending
Links
- 241000724675 Hepatitis E virus Species 0.000 title description 188
- 230000003472 neutralizing effect Effects 0.000 title description 34
- 230000027455 binding Effects 0.000 claims abstract description 84
- 108090000765 processed proteins & peptides Proteins 0.000 claims abstract description 39
- 102000004196 processed proteins & peptides Human genes 0.000 claims abstract description 36
- 229920001184 polypeptide Polymers 0.000 claims abstract description 32
- 239000012634 fragment Substances 0.000 claims abstract description 28
- 239000013604 expression vector Substances 0.000 claims abstract description 21
- 208000029564 hepatitis E virus infection Diseases 0.000 claims abstract description 13
- 108700006290 Hepatitis E virus ORF2 Proteins 0.000 claims abstract description 9
- 210000004027 cell Anatomy 0.000 claims description 81
- 239000000427 antigen Substances 0.000 claims description 50
- 108091007433 antigens Proteins 0.000 claims description 45
- 102000036639 antigens Human genes 0.000 claims description 45
- 150000001413 amino acids Chemical class 0.000 claims description 42
- 230000003993 interaction Effects 0.000 claims description 17
- 108020004707 nucleic acids Proteins 0.000 claims description 16
- 102000039446 nucleic acids Human genes 0.000 claims description 16
- 150000007523 nucleic acids Chemical class 0.000 claims description 16
- 238000003745 diagnosis Methods 0.000 claims description 14
- 210000003719 b-lymphocyte Anatomy 0.000 claims description 12
- 238000001514 detection method Methods 0.000 claims description 12
- 239000008194 pharmaceutical composition Substances 0.000 claims description 12
- 238000003259 recombinant expression Methods 0.000 claims description 9
- 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 claims description 6
- 238000009007 Diagnostic Kit Methods 0.000 claims description 5
- 108010054477 Immunoglobulin Fab Fragments Proteins 0.000 claims description 5
- 102000001706 Immunoglobulin Fab Fragments Human genes 0.000 claims description 5
- 239000003795 chemical substances by application Substances 0.000 claims description 5
- 210000001744 T-lymphocyte Anatomy 0.000 claims description 4
- 210000000822 natural killer cell Anatomy 0.000 claims description 4
- 239000000546 pharmaceutical excipient Substances 0.000 claims description 3
- 239000003814 drug Substances 0.000 abstract description 4
- 108090000623 proteins and genes Proteins 0.000 description 59
- 208000015181 infectious disease Diseases 0.000 description 57
- 102000004169 proteins and genes Human genes 0.000 description 47
- 235000018102 proteins Nutrition 0.000 description 45
- 101710132601 Capsid protein Proteins 0.000 description 42
- 101710169459 Secreted protein ORF2 Proteins 0.000 description 42
- 239000002245 particle Substances 0.000 description 39
- DCXYFEDJOCDNAF-UHFFFAOYSA-N Asparagine Natural products OC(=O)C(N)CC(N)=O DCXYFEDJOCDNAF-UHFFFAOYSA-N 0.000 description 36
- 235000001014 amino acid Nutrition 0.000 description 35
- 229940024606 amino acid Drugs 0.000 description 34
- 238000006386 neutralization reaction Methods 0.000 description 29
- 239000000539 dimer Substances 0.000 description 27
- 230000003612 virological effect Effects 0.000 description 27
- 230000002458 infectious effect Effects 0.000 description 26
- 102100035360 Cerebellar degeneration-related antigen 1 Human genes 0.000 description 25
- 238000002965 ELISA Methods 0.000 description 25
- 239000000523 sample Substances 0.000 description 23
- 108010047041 Complementarity Determining Regions Proteins 0.000 description 22
- 241000700605 Viruses Species 0.000 description 20
- 239000013078 crystal Substances 0.000 description 19
- 230000003834 intracellular effect Effects 0.000 description 18
- 238000000034 method Methods 0.000 description 17
- 125000003275 alpha amino acid group Chemical group 0.000 description 16
- 239000000872 buffer Substances 0.000 description 16
- 238000003556 assay Methods 0.000 description 15
- 238000010494 dissociation reaction Methods 0.000 description 15
- 230000005593 dissociations Effects 0.000 description 15
- 101710159752 Poly(3-hydroxyalkanoate) polymerase subunit PhaE Proteins 0.000 description 14
- 101710130262 Probable Vpr-like protein Proteins 0.000 description 14
- 241000700159 Rattus Species 0.000 description 14
- 230000000903 blocking effect Effects 0.000 description 13
- 238000004458 analytical method Methods 0.000 description 12
- 238000002425 crystallisation Methods 0.000 description 12
- 230000008025 crystallization Effects 0.000 description 12
- 230000003389 potentiating effect Effects 0.000 description 12
- 238000000746 purification Methods 0.000 description 12
- 238000001542 size-exclusion chromatography Methods 0.000 description 12
- 108010013369 Enteropeptidase Proteins 0.000 description 11
- 150000004676 glycans Chemical class 0.000 description 11
- 239000013612 plasmid Substances 0.000 description 11
- 238000001890 transfection Methods 0.000 description 11
- 102100029727 Enteropeptidase Human genes 0.000 description 10
- 230000000890 antigenic effect Effects 0.000 description 10
- 238000011282 treatment Methods 0.000 description 10
- 241000699666 Mus <mouse, genus> Species 0.000 description 9
- 239000013598 vector Substances 0.000 description 9
- 108020003175 receptors Proteins 0.000 description 8
- 102000005962 receptors Human genes 0.000 description 8
- 108010021625 Immunoglobulin Fragments Proteins 0.000 description 7
- 102000008394 Immunoglobulin Fragments Human genes 0.000 description 7
- 108010076504 Protein Sorting Signals Proteins 0.000 description 7
- 125000000539 amino acid group Chemical group 0.000 description 7
- 239000001257 hydrogen Substances 0.000 description 7
- 229910052739 hydrogen Inorganic materials 0.000 description 7
- 239000000203 mixture Substances 0.000 description 7
- 230000001225 therapeutic effect Effects 0.000 description 7
- 108090000565 Capsid Proteins Proteins 0.000 description 6
- 102100023321 Ceruloplasmin Human genes 0.000 description 6
- 108020004414 DNA Proteins 0.000 description 6
- 241000255581 Drosophila <fruit fly, genus> Species 0.000 description 6
- LYCAIKOWRPUZTN-UHFFFAOYSA-N Ethylene glycol Chemical compound OCCO LYCAIKOWRPUZTN-UHFFFAOYSA-N 0.000 description 6
- 108010001267 Protein Subunits Proteins 0.000 description 6
- 102000002067 Protein Subunits Human genes 0.000 description 6
- HEMHJVSKTPXQMS-UHFFFAOYSA-M Sodium hydroxide Chemical compound [OH-].[Na+] HEMHJVSKTPXQMS-UHFFFAOYSA-M 0.000 description 6
- 238000010367 cloning Methods 0.000 description 6
- 238000002474 experimental method Methods 0.000 description 6
- 210000004602 germ cell Anatomy 0.000 description 6
- 201000010284 hepatitis E Diseases 0.000 description 6
- 230000001717 pathogenic effect Effects 0.000 description 6
- 238000011321 prophylaxis Methods 0.000 description 6
- 230000009257 reactivity Effects 0.000 description 6
- 238000011160 research Methods 0.000 description 6
- 210000002966 serum Anatomy 0.000 description 6
- 239000000243 solution Substances 0.000 description 6
- 210000002845 virion Anatomy 0.000 description 6
- 108010011170 Ala-Trp-Arg-His-Pro-Gln-Phe-Gly-Gly Proteins 0.000 description 5
- 239000003153 chemical reaction reagent Substances 0.000 description 5
- 230000029087 digestion Effects 0.000 description 5
- 238000006471 dimerization reaction Methods 0.000 description 5
- 238000010828 elution Methods 0.000 description 5
- 230000001524 infective effect Effects 0.000 description 5
- 238000002415 sodium dodecyl sulfate polyacrylamide gel electrophoresis Methods 0.000 description 5
- 238000006467 substitution reaction Methods 0.000 description 5
- 108060003951 Immunoglobulin Proteins 0.000 description 4
- 230000004988 N-glycosylation Effects 0.000 description 4
- 108700026244 Open Reading Frames Proteins 0.000 description 4
- 108010003723 Single-Domain Antibodies Proteins 0.000 description 4
- 239000012505 Superdex™ Substances 0.000 description 4
- 108700005078 Synthetic Genes Proteins 0.000 description 4
- 210000004369 blood Anatomy 0.000 description 4
- 239000008280 blood Substances 0.000 description 4
- 238000006243 chemical reaction Methods 0.000 description 4
- 238000003776 cleavage reaction Methods 0.000 description 4
- 201000010099 disease Diseases 0.000 description 4
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 4
- 230000000694 effects Effects 0.000 description 4
- 238000005516 engineering process Methods 0.000 description 4
- 230000013595 glycosylation Effects 0.000 description 4
- 238000006206 glycosylation reaction Methods 0.000 description 4
- 210000005260 human cell Anatomy 0.000 description 4
- 102000018358 immunoglobulin Human genes 0.000 description 4
- 238000011534 incubation Methods 0.000 description 4
- 210000001806 memory b lymphocyte Anatomy 0.000 description 4
- 210000003819 peripheral blood mononuclear cell Anatomy 0.000 description 4
- 230000002265 prevention Effects 0.000 description 4
- 230000004044 response Effects 0.000 description 4
- 230000007017 scission Effects 0.000 description 4
- 230000008685 targeting Effects 0.000 description 4
- 241000283707 Capra Species 0.000 description 3
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N Dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 3
- 238000012286 ELISA Assay Methods 0.000 description 3
- DHMQDGOQFOQNFH-UHFFFAOYSA-N Glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 description 3
- 241000282412 Homo Species 0.000 description 3
- 206010061598 Immunodeficiency Diseases 0.000 description 3
- 108010038807 Oligopeptides Proteins 0.000 description 3
- 102000015636 Oligopeptides Human genes 0.000 description 3
- 241000283973 Oryctolagus cuniculus Species 0.000 description 3
- 241000404883 Pisa Species 0.000 description 3
- 230000010799 Receptor Interactions Effects 0.000 description 3
- 108010076818 TEV protease Proteins 0.000 description 3
- 208000036142 Viral infection Diseases 0.000 description 3
- 230000001154 acute effect Effects 0.000 description 3
- 230000009830 antibody antigen interaction Effects 0.000 description 3
- 230000009918 complex formation Effects 0.000 description 3
- 238000013480 data collection Methods 0.000 description 3
- 102000034287 fluorescent proteins Human genes 0.000 description 3
- 108091006047 fluorescent proteins Proteins 0.000 description 3
- 206010073071 hepatocellular carcinoma Diseases 0.000 description 3
- 230000002209 hydrophobic effect Effects 0.000 description 3
- 230000000984 immunochemical effect Effects 0.000 description 3
- 230000028802 immunoglobulin-mediated neutralization Effects 0.000 description 3
- 238000001727 in vivo Methods 0.000 description 3
- 239000003112 inhibitor Substances 0.000 description 3
- 238000002347 injection Methods 0.000 description 3
- 239000007924 injection Substances 0.000 description 3
- 230000014759 maintenance of location Effects 0.000 description 3
- 230000007246 mechanism Effects 0.000 description 3
- 239000002609 medium Substances 0.000 description 3
- 238000002156 mixing Methods 0.000 description 3
- 239000000047 product Substances 0.000 description 3
- 238000012216 screening Methods 0.000 description 3
- 238000012163 sequencing technique Methods 0.000 description 3
- 238000010186 staining Methods 0.000 description 3
- 238000002198 surface plasmon resonance spectroscopy Methods 0.000 description 3
- 208000024891 symptom Diseases 0.000 description 3
- 238000002560 therapeutic procedure Methods 0.000 description 3
- 229960005486 vaccine Drugs 0.000 description 3
- 230000009385 viral infection Effects 0.000 description 3
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 2
- HZAXFHJVJLSVMW-UHFFFAOYSA-N 2-Aminoethan-1-ol Chemical compound NCCO HZAXFHJVJLSVMW-UHFFFAOYSA-N 0.000 description 2
- JKMHFZQWWAIEOD-UHFFFAOYSA-N 2-[4-(2-hydroxyethyl)piperazin-1-yl]ethanesulfonic acid Chemical compound OCC[NH+]1CCN(CCS([O-])(=O)=O)CC1 JKMHFZQWWAIEOD-UHFFFAOYSA-N 0.000 description 2
- 101000768957 Acholeplasma phage L2 Uncharacterized 37.2 kDa protein Proteins 0.000 description 2
- 101000823746 Acidianus ambivalens Uncharacterized 17.7 kDa protein in bps2 3'region Proteins 0.000 description 2
- 101000916369 Acidianus ambivalens Uncharacterized protein in sor 5'region Proteins 0.000 description 2
- 101000769342 Acinetobacter guillouiae Uncharacterized protein in rpoN-murA intergenic region Proteins 0.000 description 2
- 101000823696 Actinobacillus pleuropneumoniae Uncharacterized glycosyltransferase in aroQ 3'region Proteins 0.000 description 2
- 101000786513 Agrobacterium tumefaciens (strain 15955) Uncharacterized protein outside the virF region Proteins 0.000 description 2
- 101000618005 Alkalihalobacillus pseudofirmus (strain ATCC BAA-2126 / JCM 17055 / OF4) Uncharacterized protein BpOF4_00885 Proteins 0.000 description 2
- 102100020724 Ankyrin repeat, SAM and basic leucine zipper domain-containing protein 1 Human genes 0.000 description 2
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 2
- 101000967489 Azorhizobium caulinodans (strain ATCC 43989 / DSM 5975 / JCM 20966 / LMG 6465 / NBRC 14845 / NCIMB 13405 / ORS 571) Uncharacterized protein AZC_3924 Proteins 0.000 description 2
- 101000977023 Azospirillum brasilense Uncharacterized 17.8 kDa protein in nodG 5'region Proteins 0.000 description 2
- 101000823761 Bacillus licheniformis Uncharacterized 9.4 kDa protein in flaL 3'region Proteins 0.000 description 2
- 101000819719 Bacillus methanolicus Uncharacterized N-acetyltransferase in lysA 3'region Proteins 0.000 description 2
- 101000789586 Bacillus subtilis (strain 168) UPF0702 transmembrane protein YkjA Proteins 0.000 description 2
- 101000792624 Bacillus subtilis (strain 168) Uncharacterized protein YbxH Proteins 0.000 description 2
- 101000790792 Bacillus subtilis (strain 168) Uncharacterized protein YckC Proteins 0.000 description 2
- 101000819705 Bacillus subtilis (strain 168) Uncharacterized protein YlxR Proteins 0.000 description 2
- 101000948218 Bacillus subtilis (strain 168) Uncharacterized protein YtxJ Proteins 0.000 description 2
- 101000961984 Bacillus thuringiensis Uncharacterized 30.3 kDa protein Proteins 0.000 description 2
- 101000718627 Bacillus thuringiensis subsp. kurstaki Putative RNA polymerase sigma-G factor Proteins 0.000 description 2
- 101000641200 Bombyx mori densovirus Putative non-structural protein Proteins 0.000 description 2
- 108010019670 Chimeric Antigen Receptors Proteins 0.000 description 2
- 101000947633 Claviceps purpurea Uncharacterized 13.8 kDa protein Proteins 0.000 description 2
- 102100025698 Cytosolic carboxypeptidase 4 Human genes 0.000 description 2
- 101000644901 Drosophila melanogaster Putative 115 kDa protein in type-1 retrotransposable element R1DM Proteins 0.000 description 2
- 101000747702 Enterobacteria phage N4 Uncharacterized protein Gp2 Proteins 0.000 description 2
- 101000948901 Enterobacteria phage T4 Uncharacterized 16.0 kDa protein in segB-ipI intergenic region Proteins 0.000 description 2
- 101000805958 Equine herpesvirus 4 (strain 1942) Virion protein US10 homolog Proteins 0.000 description 2
- 101000790442 Escherichia coli Insertion element IS2 uncharacterized 11.1 kDa protein Proteins 0.000 description 2
- 101000758599 Escherichia coli Uncharacterized 14.7 kDa protein Proteins 0.000 description 2
- 101000788354 Escherichia phage P2 Uncharacterized 8.2 kDa protein in gpA 5'region Proteins 0.000 description 2
- 101000770304 Frankia alni UPF0460 protein in nifX-nifW intergenic region Proteins 0.000 description 2
- 101000797344 Geobacillus stearothermophilus Putative tRNA (cytidine(34)-2'-O)-methyltransferase Proteins 0.000 description 2
- 101000748410 Geobacillus stearothermophilus Uncharacterized protein in fumA 3'region Proteins 0.000 description 2
- 239000007995 HEPES buffer Substances 0.000 description 2
- 101000772675 Haemophilus influenzae (strain ATCC 51907 / DSM 11121 / KW20 / Rd) UPF0438 protein HI_0847 Proteins 0.000 description 2
- 101000631019 Haemophilus influenzae (strain ATCC 51907 / DSM 11121 / KW20 / Rd) Uncharacterized protein HI_0350 Proteins 0.000 description 2
- 101000768938 Haemophilus phage HP1 (strain HP1c1) Uncharacterized 8.9 kDa protein in int-C1 intergenic region Proteins 0.000 description 2
- 101900111963 Hepatitis E virus Capsid protein Proteins 0.000 description 2
- 101000800023 Homo sapiens 4F2 cell-surface antigen heavy chain Proteins 0.000 description 2
- 101000785414 Homo sapiens Ankyrin repeat, SAM and basic leucine zipper domain-containing protein 1 Proteins 0.000 description 2
- 101000932590 Homo sapiens Cytosolic carboxypeptidase 4 Proteins 0.000 description 2
- 101000833492 Homo sapiens Jouberin Proteins 0.000 description 2
- 101000651236 Homo sapiens NCK-interacting protein with SH3 domain Proteins 0.000 description 2
- 108010001336 Horseradish Peroxidase Proteins 0.000 description 2
- 108010067060 Immunoglobulin Variable Region Proteins 0.000 description 2
- 102000017727 Immunoglobulin Variable Region Human genes 0.000 description 2
- 102100024407 Jouberin Human genes 0.000 description 2
- 101000782488 Junonia coenia densovirus (isolate pBRJ/1990) Putative non-structural protein NS2 Proteins 0.000 description 2
- 101000811523 Klebsiella pneumoniae Uncharacterized 55.8 kDa protein in cps region Proteins 0.000 description 2
- 101000768930 Lactococcus lactis subsp. cremoris Uncharacterized protein in pepC 5'region Proteins 0.000 description 2
- 101000818409 Lactococcus lactis subsp. lactis Uncharacterized HTH-type transcriptional regulator in lacX 3'region Proteins 0.000 description 2
- 101000878851 Leptolyngbya boryana Putative Fe(2+) transport protein A Proteins 0.000 description 2
- 101000976302 Leptospira interrogans Uncharacterized protein in sph 3'region Proteins 0.000 description 2
- 101000778886 Leptospira interrogans serogroup Icterohaemorrhagiae serovar Lai (strain 56601) Uncharacterized protein LA_2151 Proteins 0.000 description 2
- 101000758828 Methanosarcina barkeri (strain Fusaro / DSM 804) Uncharacterized protein Mbar_A1602 Proteins 0.000 description 2
- 101000768804 Micromonospora olivasterospora Uncharacterized 10.9 kDa protein in fmrO 5'region Proteins 0.000 description 2
- 101001122401 Middle East respiratory syndrome-related coronavirus (isolate United Kingdom/H123990006/2012) Non-structural protein ORF3 Proteins 0.000 description 2
- 101001033003 Mus musculus Granzyme F Proteins 0.000 description 2
- 101001055788 Mycolicibacterium smegmatis (strain ATCC 700084 / mc(2)155) Pentapeptide repeat protein MfpA Proteins 0.000 description 2
- HRNLUBSXIHFDHP-UHFFFAOYSA-N N-(2-aminophenyl)-4-[[[4-(3-pyridinyl)-2-pyrimidinyl]amino]methyl]benzamide Chemical compound NC1=CC=CC=C1NC(=O)C(C=C1)=CC=C1CNC1=NC=CC(C=2C=NC=CC=2)=N1 HRNLUBSXIHFDHP-UHFFFAOYSA-N 0.000 description 2
- 108091028043 Nucleic acid sequence Proteins 0.000 description 2
- 101000740670 Orgyia pseudotsugata multicapsid polyhedrosis virus Protein C42 Proteins 0.000 description 2
- 101000769182 Photorhabdus luminescens Uncharacterized protein in pnp 3'region Proteins 0.000 description 2
- 229920001213 Polysorbate 20 Polymers 0.000 description 2
- 101000961392 Pseudescherichia vulneris Uncharacterized 29.9 kDa protein in crtE 3'region Proteins 0.000 description 2
- 101000731030 Pseudomonas oleovorans Poly(3-hydroxyalkanoate) polymerase 2 Proteins 0.000 description 2
- 101001065485 Pseudomonas putida Probable fatty acid methyltransferase Proteins 0.000 description 2
- 101000711023 Rhizobium leguminosarum bv. trifolii Uncharacterized protein in tfuA 3'region Proteins 0.000 description 2
- 101000948156 Rhodococcus erythropolis Uncharacterized 47.3 kDa protein in thcA 5'region Proteins 0.000 description 2
- 101000917565 Rhodococcus fascians Uncharacterized 33.6 kDa protein in fasciation locus Proteins 0.000 description 2
- 101001121571 Rice tungro bacilliform virus (isolate Philippines) Protein P2 Proteins 0.000 description 2
- 101000790284 Saimiriine herpesvirus 2 (strain 488) Uncharacterized 9.5 kDa protein in DHFR 3'region Proteins 0.000 description 2
- 101000818098 Spirochaeta aurantia Uncharacterized protein in trpE 3'region Proteins 0.000 description 2
- 101000936719 Streptococcus gordonii Accessory Sec system protein Asp3 Proteins 0.000 description 2
- 101001026590 Streptomyces cinnamonensis Putative polyketide beta-ketoacyl synthase 2 Proteins 0.000 description 2
- 101000788499 Streptomyces coelicolor Uncharacterized oxidoreductase in mprA 5'region Proteins 0.000 description 2
- 101001102841 Streptomyces griseus Purine nucleoside phosphorylase ORF3 Proteins 0.000 description 2
- 101000708557 Streptomyces lincolnensis Uncharacterized 17.2 kDa protein in melC2-rnhH intergenic region Proteins 0.000 description 2
- QAOWNCQODCNURD-UHFFFAOYSA-N Sulfuric acid Chemical compound OS(O)(=O)=O QAOWNCQODCNURD-UHFFFAOYSA-N 0.000 description 2
- 241000282898 Sus scrofa Species 0.000 description 2
- 101000750896 Synechococcus elongatus (strain PCC 7942 / FACHB-805) Uncharacterized protein Synpcc7942_2318 Proteins 0.000 description 2
- 101000649826 Thermotoga neapolitana Putative anti-sigma factor antagonist TM1081 homolog Proteins 0.000 description 2
- 101000827562 Vibrio alginolyticus Uncharacterized protein in proC 3'region Proteins 0.000 description 2
- 101000778915 Vibrio parahaemolyticus serotype O3:K6 (strain RIMD 2210633) Uncharacterized membrane protein VP2115 Proteins 0.000 description 2
- 101000916321 Xenopus laevis Transposon TX1 uncharacterized 149 kDa protein Proteins 0.000 description 2
- 101000760088 Zymomonas mobilis subsp. mobilis (strain ATCC 10988 / DSM 424 / LMG 404 / NCIMB 8938 / NRRL B-806 / ZM1) 20.9 kDa protein Proteins 0.000 description 2
- 238000002835 absorbance Methods 0.000 description 2
- 230000009880 aromatic-aromatic interaction Effects 0.000 description 2
- 229960001230 asparagine Drugs 0.000 description 2
- 235000009582 asparagine Nutrition 0.000 description 2
- 125000000613 asparagine group Chemical group N[C@@H](CC(N)=O)C(=O)* 0.000 description 2
- 230000000712 assembly Effects 0.000 description 2
- 238000000429 assembly Methods 0.000 description 2
- 230000008840 asymmetric binding Effects 0.000 description 2
- 239000012472 biological sample Substances 0.000 description 2
- 230000015572 biosynthetic process Effects 0.000 description 2
- 210000000234 capsid Anatomy 0.000 description 2
- 230000001684 chronic effect Effects 0.000 description 2
- 150000001875 compounds Chemical class 0.000 description 2
- 238000002447 crystallographic data Methods 0.000 description 2
- 239000012228 culture supernatant Substances 0.000 description 2
- 238000013461 design Methods 0.000 description 2
- 238000011161 development Methods 0.000 description 2
- 230000018109 developmental process Effects 0.000 description 2
- 238000009792 diffusion process Methods 0.000 description 2
- 238000009826 distribution Methods 0.000 description 2
- 231100000673 dose–response relationship Toxicity 0.000 description 2
- RDYMFSUJUZBWLH-UHFFFAOYSA-N endosulfan Chemical compound C12COS(=O)OCC2C2(Cl)C(Cl)=C(Cl)C1(Cl)C2(Cl)Cl RDYMFSUJUZBWLH-UHFFFAOYSA-N 0.000 description 2
- 239000003623 enhancer Substances 0.000 description 2
- 210000003527 eukaryotic cell Anatomy 0.000 description 2
- 239000012530 fluid Substances 0.000 description 2
- 238000002523 gelfiltration Methods 0.000 description 2
- 230000002068 genetic effect Effects 0.000 description 2
- 230000001900 immune effect Effects 0.000 description 2
- 238000003018 immunoassay Methods 0.000 description 2
- 230000005847 immunogenicity Effects 0.000 description 2
- 238000000099 in vitro assay Methods 0.000 description 2
- 239000003446 ligand Substances 0.000 description 2
- 238000002826 magnetic-activated cell sorting Methods 0.000 description 2
- 210000004962 mammalian cell Anatomy 0.000 description 2
- 238000004519 manufacturing process Methods 0.000 description 2
- 239000000463 material Substances 0.000 description 2
- 238000005259 measurement Methods 0.000 description 2
- 230000004048 modification Effects 0.000 description 2
- 238000012986 modification Methods 0.000 description 2
- 238000012544 monitoring process Methods 0.000 description 2
- 239000012452 mother liquor Substances 0.000 description 2
- 230000003287 optical effect Effects 0.000 description 2
- 244000052769 pathogen Species 0.000 description 2
- YBYRMVIVWMBXKQ-UHFFFAOYSA-N phenylmethanesulfonyl fluoride Chemical compound FS(=O)(=O)CC1=CC=CC=C1 YBYRMVIVWMBXKQ-UHFFFAOYSA-N 0.000 description 2
- 229920000642 polymer Polymers 0.000 description 2
- 239000000256 polyoxyethylene sorbitan monolaurate Substances 0.000 description 2
- 235000010486 polyoxyethylene sorbitan monolaurate Nutrition 0.000 description 2
- 230000000069 prophylactic effect Effects 0.000 description 2
- 230000006916 protein interaction Effects 0.000 description 2
- RXWNCPJZOCPEPQ-NVWDDTSBSA-N puromycin Chemical compound C1=CC(OC)=CC=C1C[C@H](N)C(=O)N[C@H]1[C@@H](O)[C@H](N2C3=NC=NC(=C3N=C2)N(C)C)O[C@@H]1CO RXWNCPJZOCPEPQ-NVWDDTSBSA-N 0.000 description 2
- 210000003296 saliva Anatomy 0.000 description 2
- 230000028327 secretion Effects 0.000 description 2
- 230000035945 sensitivity Effects 0.000 description 2
- 238000000926 separation method Methods 0.000 description 2
- 238000002864 sequence alignment Methods 0.000 description 2
- 230000000392 somatic effect Effects 0.000 description 2
- 241000894007 species Species 0.000 description 2
- 239000006228 supernatant Substances 0.000 description 2
- 238000004448 titration Methods 0.000 description 2
- 230000001052 transient effect Effects 0.000 description 2
- 210000002700 urine Anatomy 0.000 description 2
- 238000010200 validation analysis Methods 0.000 description 2
- 230000035899 viability Effects 0.000 description 2
- 239000013603 viral vector Substances 0.000 description 2
- 125000000979 2-amino-2-oxoethyl group Chemical group [H]C([*])([H])C(=O)N([H])[H] 0.000 description 1
- ZBMRKNMTMPPMMK-UHFFFAOYSA-N 2-amino-4-[hydroxy(methyl)phosphoryl]butanoic acid;azane Chemical compound [NH4+].CP(O)(=O)CCC(N)C([O-])=O ZBMRKNMTMPPMMK-UHFFFAOYSA-N 0.000 description 1
- UAIUNKRWKOVEES-UHFFFAOYSA-N 3,3',5,5'-tetramethylbenzidine Chemical compound CC1=C(N)C(C)=CC(C=2C=C(C)C(N)=C(C)C=2)=C1 UAIUNKRWKOVEES-UHFFFAOYSA-N 0.000 description 1
- 102100033400 4F2 cell-surface antigen heavy chain Human genes 0.000 description 1
- 206010069754 Acquired gene mutation Diseases 0.000 description 1
- 241000251468 Actinopterygii Species 0.000 description 1
- 239000012103 Alexa Fluor 488 Substances 0.000 description 1
- 239000012117 Alexa Fluor 700 Substances 0.000 description 1
- 108010032595 Antibody Binding Sites Proteins 0.000 description 1
- 206010003445 Ascites Diseases 0.000 description 1
- 241000228212 Aspergillus Species 0.000 description 1
- 108091008875 B cell receptors Proteins 0.000 description 1
- 102100024222 B-lymphocyte antigen CD19 Human genes 0.000 description 1
- 241000193830 Bacillus <bacterium> Species 0.000 description 1
- 241000702009 Bacillus phage SPR Species 0.000 description 1
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 1
- 101100454807 Caenorhabditis elegans lgg-1 gene Proteins 0.000 description 1
- 241000282832 Camelidae Species 0.000 description 1
- 101710167800 Capsid assembly scaffolding protein Proteins 0.000 description 1
- 241001466804 Carnivora Species 0.000 description 1
- 102000053642 Catalytic RNA Human genes 0.000 description 1
- 108090000994 Catalytic RNA Proteins 0.000 description 1
- 241000282994 Cervidae Species 0.000 description 1
- 241000288673 Chiroptera Species 0.000 description 1
- 108020004705 Codon Proteins 0.000 description 1
- 208000035473 Communicable disease Diseases 0.000 description 1
- 102100027819 Cytosolic beta-glucosidase Human genes 0.000 description 1
- 108090000626 DNA-directed RNA polymerases Proteins 0.000 description 1
- 102000004163 DNA-directed RNA polymerases Human genes 0.000 description 1
- 108700022810 Drosophila Hsc70-3 Proteins 0.000 description 1
- 241000255601 Drosophila melanogaster Species 0.000 description 1
- 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 1
- 208000005189 Embolism Diseases 0.000 description 1
- 241000196324 Embryophyta Species 0.000 description 1
- 241000588724 Escherichia coli Species 0.000 description 1
- 108010087819 Fc receptors Proteins 0.000 description 1
- 102000009109 Fc receptors Human genes 0.000 description 1
- 102000002090 Fibronectin type III Human genes 0.000 description 1
- 108050009401 Fibronectin type III Proteins 0.000 description 1
- 239000004471 Glycine Substances 0.000 description 1
- 102000003886 Glycoproteins Human genes 0.000 description 1
- 108090000288 Glycoproteins Proteins 0.000 description 1
- 241000711549 Hepacivirus C Species 0.000 description 1
- 208000037319 Hepatitis infectious Diseases 0.000 description 1
- 206010019799 Hepatitis viral Diseases 0.000 description 1
- 241000238631 Hexapoda Species 0.000 description 1
- 101000980825 Homo sapiens B-lymphocyte antigen CD19 Proteins 0.000 description 1
- 101000859692 Homo sapiens Cytosolic beta-glucosidase Proteins 0.000 description 1
- 101000997662 Homo sapiens Lysosomal acid glucosylceramidase Proteins 0.000 description 1
- 102000014150 Interferons Human genes 0.000 description 1
- 108010050904 Interferons Proteins 0.000 description 1
- 241000124008 Mammalia Species 0.000 description 1
- 241001465754 Metazoa Species 0.000 description 1
- 108060004795 Methyltransferase Proteins 0.000 description 1
- 239000012901 Milli-Q water Substances 0.000 description 1
- 241000699670 Mus sp. Species 0.000 description 1
- 101710152005 Non-structural polyprotein Proteins 0.000 description 1
- 101710113540 ORF2 protein Proteins 0.000 description 1
- 239000012124 Opti-MEM Substances 0.000 description 1
- 240000007019 Oxalis corniculata Species 0.000 description 1
- 229930040373 Paraformaldehyde Natural products 0.000 description 1
- 208000005228 Pericardial Effusion Diseases 0.000 description 1
- 208000037581 Persistent Infection Diseases 0.000 description 1
- NBIIXXVUZAFLBC-UHFFFAOYSA-N Phosphoric acid Chemical compound OP(O)(O)=O NBIIXXVUZAFLBC-UHFFFAOYSA-N 0.000 description 1
- 101710114167 Polyprotein P1234 Proteins 0.000 description 1
- 101710124590 Polyprotein nsP1234 Proteins 0.000 description 1
- 108010002519 Prolactin Receptors Proteins 0.000 description 1
- 102100029000 Prolactin receptor Human genes 0.000 description 1
- 101710090523 Putative movement protein Proteins 0.000 description 1
- 238000003559 RNA-seq method Methods 0.000 description 1
- 241000235342 Saccharomycetes Species 0.000 description 1
- VMHLLURERBWHNL-UHFFFAOYSA-M Sodium acetate Chemical compound [Na+].CC([O-])=O VMHLLURERBWHNL-UHFFFAOYSA-M 0.000 description 1
- 108091081024 Start codon Proteins 0.000 description 1
- 241000187747 Streptomyces Species 0.000 description 1
- 101710120037 Toxin CcdB Proteins 0.000 description 1
- 239000007983 Tris buffer Substances 0.000 description 1
- 239000013504 Triton X-100 Substances 0.000 description 1
- 229920004890 Triton X-100 Polymers 0.000 description 1
- GLNADSQYFUSGOU-GPTZEZBUSA-J Trypan blue Chemical compound [Na+].[Na+].[Na+].[Na+].C1=C(S([O-])(=O)=O)C=C2C=C(S([O-])(=O)=O)C(/N=N/C3=CC=C(C=C3C)C=3C=C(C(=CC=3)\N=N\C=3C(=CC4=CC(=CC(N)=C4C=3O)S([O-])(=O)=O)S([O-])(=O)=O)C)=C(O)C2=C1N GLNADSQYFUSGOU-GPTZEZBUSA-J 0.000 description 1
- 102000018265 Virus Receptors Human genes 0.000 description 1
- 108010066342 Virus Receptors Proteins 0.000 description 1
- 230000002745 absorbent Effects 0.000 description 1
- 239000002250 absorbent Substances 0.000 description 1
- 230000021736 acetylation Effects 0.000 description 1
- 238000006640 acetylation reaction Methods 0.000 description 1
- 230000004913 activation Effects 0.000 description 1
- 239000004480 active ingredient Substances 0.000 description 1
- 230000006978 adaptation Effects 0.000 description 1
- 238000001261 affinity purification Methods 0.000 description 1
- 238000007818 agglutination assay Methods 0.000 description 1
- 238000012867 alanine scanning Methods 0.000 description 1
- 150000001412 amines Chemical class 0.000 description 1
- 210000004381 amniotic fluid Anatomy 0.000 description 1
- 239000012491 analyte Substances 0.000 description 1
- 230000019552 anatomical structure morphogenesis Effects 0.000 description 1
- 230000003466 anti-cipated effect Effects 0.000 description 1
- 230000000692 anti-sense effect Effects 0.000 description 1
- 230000000840 anti-viral effect Effects 0.000 description 1
- 230000009833 antibody interaction Effects 0.000 description 1
- 230000005875 antibody response Effects 0.000 description 1
- 210000000628 antibody-producing cell Anatomy 0.000 description 1
- 102000025171 antigen binding proteins Human genes 0.000 description 1
- 108091000831 antigen binding proteins Proteins 0.000 description 1
- 239000003443 antiviral agent Substances 0.000 description 1
- 210000003567 ascitic fluid Anatomy 0.000 description 1
- 230000001580 bacterial effect Effects 0.000 description 1
- 230000006399 behavior Effects 0.000 description 1
- 230000008901 benefit Effects 0.000 description 1
- 230000004071 biological effect Effects 0.000 description 1
- 238000013378 biophysical characterization Methods 0.000 description 1
- 239000007853 buffer solution Substances 0.000 description 1
- 210000004899 c-terminal region Anatomy 0.000 description 1
- 150000001720 carbohydrates Chemical group 0.000 description 1
- 239000000969 carrier Substances 0.000 description 1
- 239000013592 cell lysate Substances 0.000 description 1
- 238000005119 centrifugation Methods 0.000 description 1
- 210000001175 cerebrospinal fluid Anatomy 0.000 description 1
- 210000004978 chinese hamster ovary cell Anatomy 0.000 description 1
- 210000000349 chromosome Anatomy 0.000 description 1
- 238000004891 communication Methods 0.000 description 1
- 238000012875 competitive assay Methods 0.000 description 1
- 230000000295 complement effect Effects 0.000 description 1
- 230000003750 conditioning effect Effects 0.000 description 1
- 238000012790 confirmation Methods 0.000 description 1
- 230000009260 cross reactivity Effects 0.000 description 1
- 230000009849 deactivation Effects 0.000 description 1
- 230000034994 death Effects 0.000 description 1
- 231100000517 death Toxicity 0.000 description 1
- 230000002939 deleterious effect Effects 0.000 description 1
- 238000012217 deletion Methods 0.000 description 1
- 230000037430 deletion Effects 0.000 description 1
- 230000001419 dependent effect Effects 0.000 description 1
- 230000000368 destabilizing effect Effects 0.000 description 1
- 238000002050 diffraction method Methods 0.000 description 1
- 238000010790 dilution Methods 0.000 description 1
- 239000012895 dilution Substances 0.000 description 1
- 229940042399 direct acting antivirals protease inhibitors Drugs 0.000 description 1
- 239000003651 drinking water Substances 0.000 description 1
- 235000020188 drinking water Nutrition 0.000 description 1
- 241001493065 dsRNA viruses Species 0.000 description 1
- 238000004520 electroporation Methods 0.000 description 1
- 210000002472 endoplasmic reticulum Anatomy 0.000 description 1
- 230000017188 evasion or tolerance of host immune response Effects 0.000 description 1
- 230000007717 exclusion Effects 0.000 description 1
- 210000003608 fece Anatomy 0.000 description 1
- 239000012091 fetal bovine serum Substances 0.000 description 1
- 238000001914 filtration Methods 0.000 description 1
- 238000000684 flow cytometry Methods 0.000 description 1
- 239000012737 fresh medium Substances 0.000 description 1
- 230000005714 functional activity Effects 0.000 description 1
- 108020001507 fusion proteins Proteins 0.000 description 1
- 102000037865 fusion proteins Human genes 0.000 description 1
- 238000010353 genetic engineering Methods 0.000 description 1
- 125000003712 glycosamine group Chemical group 0.000 description 1
- 102000035122 glycosylated proteins Human genes 0.000 description 1
- 108091005608 glycosylated proteins Proteins 0.000 description 1
- 230000012010 growth Effects 0.000 description 1
- 229960000789 guanidine hydrochloride Drugs 0.000 description 1
- PJJJBBJSCAKJQF-UHFFFAOYSA-N guanidinium chloride Chemical compound [Cl-].NC(N)=[NH2+] PJJJBBJSCAKJQF-UHFFFAOYSA-N 0.000 description 1
- 230000036541 health Effects 0.000 description 1
- 108010080271 hecolin Proteins 0.000 description 1
- 208000005252 hepatitis A Diseases 0.000 description 1
- 210000003494 hepatocyte Anatomy 0.000 description 1
- HNDVDQJCIGZPNO-UHFFFAOYSA-N histidine Natural products OC(=O)C(N)CC1=CN=CN1 HNDVDQJCIGZPNO-UHFFFAOYSA-N 0.000 description 1
- 230000003053 immunization Effects 0.000 description 1
- 238000002649 immunization Methods 0.000 description 1
- 238000010166 immunofluorescence Methods 0.000 description 1
- 238000010820 immunofluorescence microscopy Methods 0.000 description 1
- 238000003125 immunofluorescent labeling Methods 0.000 description 1
- 239000002955 immunomodulating agent Substances 0.000 description 1
- 229940121354 immunomodulator Drugs 0.000 description 1
- 238000001114 immunoprecipitation Methods 0.000 description 1
- 238000000338 in vitro Methods 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
- 229940047124 interferons Drugs 0.000 description 1
- 238000011835 investigation Methods 0.000 description 1
- 238000002955 isolation Methods 0.000 description 1
- 230000000670 limiting effect Effects 0.000 description 1
- 238000012417 linear regression Methods 0.000 description 1
- 239000007788 liquid Substances 0.000 description 1
- 238000011068 loading method Methods 0.000 description 1
- 210000004072 lung Anatomy 0.000 description 1
- 210000004880 lymph fluid Anatomy 0.000 description 1
- 108010026228 mRNA guanylyltransferase Proteins 0.000 description 1
- 238000007885 magnetic separation Methods 0.000 description 1
- 238000013507 mapping Methods 0.000 description 1
- 230000035800 maturation Effects 0.000 description 1
- 239000011325 microbead Substances 0.000 description 1
- 235000013336 milk Nutrition 0.000 description 1
- 210000004080 milk Anatomy 0.000 description 1
- 239000008267 milk Substances 0.000 description 1
- 231100000350 mutagenesis Toxicity 0.000 description 1
- 230000035772 mutation Effects 0.000 description 1
- 239000013642 negative control Substances 0.000 description 1
- 238000007481 next generation sequencing Methods 0.000 description 1
- 229910052757 nitrogen Inorganic materials 0.000 description 1
- 239000013631 noncovalent dimer Substances 0.000 description 1
- 230000009871 nonspecific binding Effects 0.000 description 1
- 239000002773 nucleotide Substances 0.000 description 1
- 125000003729 nucleotide group Chemical group 0.000 description 1
- 238000005457 optimization Methods 0.000 description 1
- 230000008520 organization Effects 0.000 description 1
- 229920002866 paraformaldehyde Polymers 0.000 description 1
- 230000037361 pathway Effects 0.000 description 1
- 239000000137 peptide hydrolase inhibitor Substances 0.000 description 1
- 210000004912 pericardial fluid Anatomy 0.000 description 1
- 230000008823 permeabilization Effects 0.000 description 1
- 235000011007 phosphoric acid Nutrition 0.000 description 1
- 230000026731 phosphorylation Effects 0.000 description 1
- 238000006366 phosphorylation reaction Methods 0.000 description 1
- 235000015277 pork Nutrition 0.000 description 1
- 230000004481 post-translational protein modification Effects 0.000 description 1
- 239000002244 precipitate Substances 0.000 description 1
- 238000002360 preparation method Methods 0.000 description 1
- 238000002203 pretreatment Methods 0.000 description 1
- 230000003449 preventive effect Effects 0.000 description 1
- 210000001236 prokaryotic cell Anatomy 0.000 description 1
- 230000001681 protective effect Effects 0.000 description 1
- 238000000159 protein binding assay Methods 0.000 description 1
- 230000002797 proteolythic effect Effects 0.000 description 1
- 229950010131 puromycin Drugs 0.000 description 1
- 238000011158 quantitative evaluation Methods 0.000 description 1
- 238000002708 random mutagenesis Methods 0.000 description 1
- 239000011541 reaction mixture Substances 0.000 description 1
- 230000009467 reduction Effects 0.000 description 1
- 230000002829 reductive effect Effects 0.000 description 1
- 230000001105 regulatory effect Effects 0.000 description 1
- 230000010076 replication Effects 0.000 description 1
- 230000001177 retroviral effect Effects 0.000 description 1
- 108091092562 ribozyme Proteins 0.000 description 1
- 239000012146 running buffer Substances 0.000 description 1
- 238000003118 sandwich ELISA Methods 0.000 description 1
- 230000003248 secreting effect Effects 0.000 description 1
- 238000002741 site-directed mutagenesis Methods 0.000 description 1
- 235000020183 skimmed milk Nutrition 0.000 description 1
- 239000001632 sodium acetate Substances 0.000 description 1
- 235000017281 sodium acetate Nutrition 0.000 description 1
- 239000007787 solid Substances 0.000 description 1
- 230000037439 somatic mutation Effects 0.000 description 1
- 230000000087 stabilizing effect Effects 0.000 description 1
- 238000010561 standard procedure Methods 0.000 description 1
- 210000004243 sweat Anatomy 0.000 description 1
- 210000001179 synovial fluid Anatomy 0.000 description 1
- 238000003786 synthesis reaction Methods 0.000 description 1
- 238000010998 test method Methods 0.000 description 1
- 238000012360 testing method Methods 0.000 description 1
- 230000002103 transcriptional effect Effects 0.000 description 1
- 239000012096 transfection reagent Substances 0.000 description 1
- 238000012546 transfer Methods 0.000 description 1
- 230000009466 transformation Effects 0.000 description 1
- 230000014616 translation Effects 0.000 description 1
- 230000005945 translocation Effects 0.000 description 1
- 230000001296 transplacental effect Effects 0.000 description 1
- LENZDBCJOHFCAS-UHFFFAOYSA-N tris Chemical compound OCC(N)(CO)CO LENZDBCJOHFCAS-UHFFFAOYSA-N 0.000 description 1
- 239000003656 tris buffered saline Substances 0.000 description 1
- 201000001862 viral hepatitis Diseases 0.000 description 1
- 238000005406 washing Methods 0.000 description 1
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 1
- 238000001262 western blot Methods 0.000 description 1
- 210000005253 yeast cell Anatomy 0.000 description 1
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/08—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses
- C07K16/10—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses from RNA viruses
-
- 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
- C07K2317/30—Immunoglobulins specific features characterized by aspects of specificity or valency
- C07K2317/34—Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
-
- 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
- C07K2317/76—Antagonist effect on antigen, e.g. neutralization or inhibition of binding
-
- 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
Definitions
- the present invention relates to broadly neutralizing monoclonal antibodies against infectious hepatitis E virus particles.
- the present invention relates to antibody constructs capable of specifically binding to a conformational epitope of a non-glycosylated polypeptide encoded by hepatitis E virus ORF2, e.g., to monoclonal antibody p60.1 or monoclonal antibody p60.12 or single chains, derivatives or antigen-binding fragments thereof, as specified herein. It also provides heavy chain constructs and/or a light chain constructs of said antibody constructs.
- the antibody constructs, expression vectors encoding them and host cells capable of expressing them are useful in medicine, e.g., for preventing, treating and diagnosing hepatitis E virus infection.
- Hepatitis E virus is the most common cause of acute viral hepatitis worldwide and is an emerging problem in industrialized countries. Approximately 2 billion people live in areas endemic for HEV and are at risk of infection. It is estimated that one third of the population in the world may be infected with HEV during their lifetime. This virus infects approximately 20 million people every year and is responsible for an estimated 3.4 million symptomatic cases and 70,000 deaths, mainly in developing countries. While HEV infection is asymptomatic for most patients, some human populations including pregnant women and immunocompromised patients have higher risk to develop severe forms and chronic infections, respectively. Currently, there is neither a specific treatment nor a universal vaccine against HEV.
- HEV strains infecting humans have been classified into 4 main distinct genotypes (gt) belonging to a single serotype. Genotypes gt1 and gt2 that infect humans only, are primarily transmitted through contaminated drinking water and are responsible for waterborne hepatitis outbreaks in developing countries. In contrast, gt3 and gt4 are zoonotic and are largely circulating in industrialized countries. They are mainly transmitted by contact with swine and consumption of inadequately heated pork products. Recently, further HEV genotypes, subgenotypes and strains have been identified in rabbit, hare, wild boar, moose, camels, and other animals.
- rat HEV In addition to human pathogenic HEV, related viruses have been identified in fowl, rats, bats, carnivores and even in fish. After its initial description in Germany, rat HEV has been detected in many regions of the world suggesting a worldwide distribution. Recently, several rat HEV-caused human disease cases have been reported, and rat HEV is thought to pose a considerable zoonotic threat.
- HEV is a quasi-enveloped, positive-sense RNA virus expressing three open reading frames (ORFs): ORF1 , ORF2 and ORF3.
- ORF1 encodes the non-structural polyprotein that is the viral replicase.
- ORF2 encodes the viral capsid protein which is involved in particle assembly, binding to host cells and eliciting neutralizing antibodies.
- ORF3 encodes a small protein that is involved in virion morphogenesis and egress.
- ORF2 is the best characterized of the three ORFs. Its protein, pORF2, is the main structural component of the virion, carrying the receptor binding domain, the major target for neutralizing antibodies. Three potential N-linked glycosylation sites at positions N137, N310, and N562 have been identified in pORF2, but N-linked glycosylation has only been confirmed to occur at positions N137 and N562. During its lifecycle, HEV produces at least 3 forms of the pORF2 capsid protein: infectious pORF2 (pORF2i), glycosylated pORF2 (pORF2g), and cleaved pORF2 (pORF2c).
- the pORF2i protein is the structural component of infectious particles.
- pORF2g and pORF2c are highly glycosylated, and are not associated with the infectious particle. Notably, these secreted forms are very stable, exist as dimers, and are the most abundant forms of pORF2 in sera of HEV infected patients. These proteins probably do not play an important role in the life cycle of HEV, but may inhibit antibody mediated neutralization, thus serving as an immune evasion mechanism for HEV.
- pORF2g and pORF2c likely act as a humoral immune decoy that inhibits antibody-mediated neutralization.
- WO2018138344A1 Hepatitis E virus ORF2 capsid polypeptides and uses thereof’ concerns hepatitis E virus pORF2 polypeptides which may be both glycosylated or not and antibodies specific for either the non-glycosylated pORF2i or the glycosylated pORF2g polypeptides for use in HEV diagnosis.
- WO2018138344A1 does not teach the therapeutic use of antibodies specific for the non-glycosylated pORF2i.
- W02020011755A1 Antibodies having specificity for the ORF2i protein of hepatitis E virus and uses thereof for diagnostic purposes” concern antibodies which specifically bind to pORF2i and not to pORF2g or pORF2c, to a linear epitope around glycosylation site N3 562NTT.
- W02020011755A1 does not teach the therapeutic use of antibodies specific for the non-glycosylated pORF2i.
- detection of infectious virus particles under non-denaturing and non-reducing conditions e.g., in stool or plasma samples from HEV infected individuals
- ELISA detection of infectious virus particles under non-denaturing and non-reducing conditions (e.g., in stool or plasma samples from HEV infected individuals) by ELISA using antibodies described in W02020011755A1 has not been shown. This is likely due to the linear character of the targeted epitope described therein that is not accessible in natively folded pORF2 dimers constituting infectious particles
- the present invention in one aspect relates to a heavy chain construct and/or a light chain construct of an antibody construct capable of specifically binding to a conformational epitope of a non-glycosylated polypeptide encoded by hepatitis E virus ORF2, a) wherein the heavy chain construct comprises a CDR1 having at least 90% sequence identity to SEQ ID NO: 2, a CDR2 having at least 88% sequence identity to SEQ ID NO: 3, and a CDR3 having at least 90% sequence identity to SEQ ID NO: 4, and/or the light chain construct comprises a CDR1 having at least 83% sequence identity to SEQ ID NO: 6, a CDR2 having at least 66% sequence identity to the sequence AAS, and a CDR3 having at least 85% sequence identity to SEQ ID NO: 8; or b) wherein the heavy chain construct comprises a CDR1 having at least 87% sequence identity to SEQ ID NO: 10, a CDR2 having at least 87% sequence identity to SEQ ID NO: 11 , and a CDR3
- said heavy chain construct and/or light chain construct of the invention comprise a) a heavy chain variable region having at least 90% sequence identity to amino acids 1-
- the antibody construct may be a Fab fragment, a Fab 2 fragment or an scFv or a single chain antibody.
- the antibody construct may be a human antibody, optionally, a human antibody comprising a constant region.
- sequence identity in the CDR1 , CDR2, and/or CDR3 may be 100% (to the respective defined CDRs), wherein, preferably, all sequence identity in the CDRs is 100%.
- sequence identity in the variable regions also is 100% to the respective defined variable regions.
- the invention provides an antibody construct comprising the heavy chain construct and light chain construct of the invention, i.e. , an antibody construct capable of specifically binding to a conformational epitope of a non-glycosylated polypeptide encoded by hepatitis E virus ORF2.
- the present invention also relates to a nucleic acid molecule encoding a heavy chain construct and/or light chain construct of the invention or an antibody construct of the invention, as well as to a recombinant expression vector comprising said nucleic acid molecule, preferably, under the control of a heterologous promoter, and to a host cell comprising said recombinant expression vector, which is able to express the heavy chain construct and/or light chain construct or the antibody construct of the invention.
- the cell may be, e.g., a B cell, a T cell or an NK cell.
- the present invention also relates to said antibody construct for use in the diagnosis of hepatitis E virus infection.
- the present invention also relates to a diagnostic kit for the diagnosis of hepatitis E virus infection, comprising at least one of the antibody constructs of the invention, , or any combination thereof, in an amount effective for diagnosis, and, optionally, detection agents suitable for detection of the interaction of antigen and antibody construct.
- the present invention also relates to the use of said antibody constructs as a medicament for prophylaxis and/or treatment of hepatitis E virus infection.
- composition comprising a) the antibody construct of the invention, b) the expression vector of the invention, wherein the nucleic acid encodes the antibody construct of the invention and/or c) the host cell of the invention, wherein the nucleic acid encodes the antibody construct of the invention, and, optionally, a pharmaceutically acceptable vehicle and/or excipient.
- the present invention also relates to said pharmaceutical composition for use in preventing or treating hepatitis E virus infection.
- FIG 1 is a schematic illustration of HEV pORF2 domains (A) and expression constructs for the P domain fused to either mNeon or mRuby (B).
- pGS38 intracellular P domain
- pGS39 secreted P domain.
- the secreted construct carried a BiP signal sequence to allow translocation into the endoplasmic reticulum. Numbers indicate amino acid positions within HEV gt3 pORF2 (SEQ ID NO: 17).
- Figure 2 shows the results of neutralization assays of HEV Kernow C1 p6 G1634R viral strain. Neutralization of naked viral particles by p60.1 (A) and p60.12 (B) as well as neutralization of pseudo-enveloped viral particles by p60.1 (C) and p60.12 (D) is shown. Each panel shows the titration results (left) and the respective dose response fitted curve used to calculate the IC50 (right). Panel E shows neutralization results of p60.1 and p60.12 against the gt3 HEV viral strain 83-2-27. FFU/well normalized to the control are depicted.
- Figure 3 illustrates monoclonal antibody expression and purification:
- A Size exclusion chromatography profiles representing the general behavior of the antibodies p60.1 and p60.12 after separation on SD200 increase 10/300 GL column equilibrated in 1x PBS at a flow rate of 0.5 ml/min.
- B A 12 % Coomassie-stained SDS-PAGE gel with the antibodies p60.1 and p60.12 loaded under reducing conditions. The estimated molecular weight for the heavy chain is ca. 55 kDa and ca. 25 kDa for the light chain.
- Panel B shows the results of binding of the depicted antibody to proteins present in hepatoma cells transfected with the indicated viral strain.
- the GLUC reporter replicon was used as negative control as it does not contain ORF2.
- convalescent patient serum was used.
- p60.1 shows binding to the gt1 viral strain SAR55.
- Figure 6 shows ELISA results using the anti-HEV monoclonal antibodies p60.1 or p60.12 together with sera from infected patients.
- the indicated antibodies were used to capture antigens present in HEV RNA positive patient plasma.
- Figure 7 illustrates the differential binding activity of the anti-HEV monoclonal antibodies of the invention as tested by size exclusion chromatography.
- the elution profiles demonstrate that scFv-p60.1 and scFv-p60.12 only bind to the non-secreted P domain (pGS99) and not to the secreted P domain (pGS100) from HEV gt3.
- Figure 8 illustrates the determination of P domain-monoclonal antibody binding properties:
- A Sensorgrams showing association and dissociation phases of the interaction between non-secreted HEV gt3 P domain and the neutralizing antibodies of the invention.
- B Sensorgrams showing association and dissociation phases of the interaction between secreted HEV gt3 P domain and the neutralizing antibodies of the invention.
- Analyte concentrations (mAbs) were 5, 10, 25, 50 and 75 nM. Binding data are shown as black lines and fitting is shown overlaid in gray lines.
- Figure 9 shows the size exclusion chromatography profiles obtained from complexes of anti-HEV scFvs with the HEV gt3 P domain for scFv-p60.1 and pGS99 and scFv-p60.12 and pGS99.
- Figure 10 shows the crystals obtained from complexes of anti-HEV scFvs with the HEV gt3 P domain (pGS99) in complex with the neutralizing mAbs (A) scFv-p60.1 and (B) Fab- p60.12.
- Figure 11 shows the crystal structure of the HEV gt3 P domain (pGS99) in complex with the neutralizing mAb scFv-p60.1 , shown in cartoon representation with the heavy (VH) and light chains (VL).
- the P domain dimer has one protomer colored black and the other grey.
- One complex in the asymmetric unit shows the P domain with surface representation, while the other is a cartoon representation.
- Figure 12 depicts an overview (top panels) and close-up view (lower panels) of antibodyantigen interfaces of p60.1 (A) and p60.12 (B) in complex with the dimeric HEV gt3 P domain.
- the antibody dasheavy chain in dark grey, light chain in light grey
- contacts the two protomers of the P domain grey, shown at the bottom
- the asparagine side chain of N562 shown in sticks
- Figure 13 shows a comparison of the complexes between the HEV gt3 P domain dimer with human (scFv-p60.1) and mouse (scFv from Fab 8C11 and scFv from Fab 8G12) anti- HEV antibodies.
- Figure 14 illustrates the fitting of the crystal structures of the HEV gt3 P domain in complex with mouse and human scFvs to HEV virus-like particle (VLP) structure.
- the VLP (PDB 2ztn) is shown as surface representation while the scFvs are shown as cartoons.
- the P domain was used to fit the complexes to the VLP.
- Figure 15 shows the amino acid sequence alignment of the P domains from the five HEV genotypes studied (SEQ ID NO: 18-22).
- Contact residues for the mAbs are indicated as follows: ⁇ : contact residues shared between p60.1 and p60.12 epitope. •: contact residues for p60.12 and ⁇ : contact residues for p60.1.
- Neutralizing antibodies can be classified into two groups, the first group comprising antibodies that inhibit only the infecting viral variant and not any other virus strains or variants. These antibodies are known as autologous antibodies and their neutralization is considered transient and highly affected by viral evolution.
- the second group of antibodies comprises broadly neutralizing antibodies (bNAbs). These have the ability to neutralize several variants of the same virus or members of the same viral family. Such antibodies often target epitopes that are highly conserved across diverse strains, virus subtypes and families.
- the present invention provides highly potent bNAbs, which may be used for the diagnosis, the prophylaxis and/or the therapy of HEV, showing high binding affinity, potent neutralization activity, as well as broad reactivity across HEV genotypes, including human and rat HEV.
- Said antibodies or antibody constructs specifically target a glycan-dependent conformational neutralization epitope in pORF2 that is only accessible in the infectious (non-glycosylated) HEV particle, so that their neutralization activity is specific for the non- glycosylated form of pORF2, and cannot be evaded by the soluble secreted (glycosylated) forms of pORF2.
- the first object of the present invention is a heavy chain construct and/or a light chain construct of an antibody construct capable of specifically binding to a conformational epitope of a non-glycosylated polypeptide encoded by hepatitis E virus ORF2, a) wherein the heavy chain construct comprises a CDR1 having at least 90% sequence identity to SEQ ID NO: 2, a CDR2 having at least 88% sequence identity to SEQ ID NO: 3, and a CDR3 having at least 90% sequence identity to SEQ ID NO: 4, and/or the light chain construct comprises a CDR1 having at least 83% sequence identity to SEQ ID NO: 6, a CDR2 having at least 66% sequence identity to the sequence AAS, and a CDR3 having at least 85% sequence identity to SEQ ID NO: 8 (i.e., the CDRs of monoclonal antibody p60.1); or b) wherein the heavy chain construct comprises a CDR1 having at least 87% sequence identity to SEQ ID NO: 10, a CDR2 having
- said heavy chain construct and/or light chain construct of the invention comprise a) a heavy chain variable region having at least 90% sequence identity to amino acids 1-
- the invention provides a heavy chain construct and/or light chain construct of the invention, wherein a) the heavy chain construct comprises a CDR1 having at least 90% sequence identity to SEQ ID NO: 2, a CDR2 having at least 88% sequence identity to SEQ ID NO: 3, and a CDR3 having at least 90% sequence identity to SEQ ID NO: 4, and/or the light chain construct comprises a CDR1 having at least 83% sequence identity to SEQ ID NO: 6, a CDR2 having at least 66% sequence identity to the sequence AAS, and a CDR3 having at least 85% sequence identity to SEQ ID NO: 8.
- the heavy chain variable region has at least 90% sequence identity to amino acids 1-121 of SEQ ID NO: 1 and/or and the light chain variable region has at least 90% sequence identity to amino acids 1-106 of SEQ ID NO: 5, and, optionally, the heavy chain has at least 90% sequence identity to SEQ ID NO: 1 and/or and light chain has at least 90% sequence identity to SEQ ID NO: 5.
- Antibody constructs comprising said CDRs, said variable regions or said heavy and light chain constructs are designated p60.1.
- the preferred scFv p.60.1 comprises a heavy chain construct of SEQ ID NO: 1 and a light chain construct of SEQ ID NO: 5.
- the invention provides a heavy chain construct and/or light chain construct of the invention, wherein a) the heavy chain construct comprises a CDR1 having at least 87% sequence identity to SEQ ID NO: 10, a CDR2 having at least 87% sequence identity to SEQ ID NO: 11 , and a CDR3 having at least 93% sequence identity to SEQ ID NO: 12 and/or the light chain construct comprises a CDR1 having at least 88% sequence identity to SEQ ID NO: 14, a CDR2 having at least 66% sequence identity to the sequence DVT, and a CDR3 having at least 90% sequence identity to SEQ ID NO: 16.
- the heavy chain variable region has at least 90% sequence identity to amino acids 1-122 of SEQ ID NO: 9 and/or and the light chain variable region has at least 90% sequence identity to amino acids 1-111 of SEQ ID NO: 13, and, optionally, the heavy chain has at least 90% sequence identity to SEQ ID NO: 9 and/or and light chain has at least 90% sequence identity to SEQ ID NO: 13.
- Antibody constructs comprising said CDRs, said variable regions or said heavy and light chain constructs are designated p60.12.
- the preferred scFv p.60.12 comprises a heavy chain construct of SEQ ID NO: 9 and a light chain construct of SEQ ID NO: 13.
- antibody construct or “antibody”, also “immunoglobulin”, has its general meaning in the art and refers to any antibody-like molecule that has an antigen binding region, and this term includes antibody fragments that comprise an antigen binding domain such as Fab', Fab, F(ab')2, and single domain antibodies (DABs). Chimeric antigen receptors (CARs) are also considered antibody constructs.
- Antibody constructs may, as in natural antibodies, e.g., of the IgG class, comprise two heavy chains linked to each other by disulfide bonds, wherein each heavy chain is linked to a light chain by a disulfide bond.
- Antibody constructs of the invention may comprise constant regions, and may belong to each isotype.
- antibodies of the invention may be of the IgG isotype, e.g. IgGi or lgG4. Each chain contains distinct sequence domains.
- the light chain typically includes two domains, a variable domain (VL) and a constant domain (CL).
- the heavy chain typically includes four domains, a variable domain (VH) and three constant domains (CH1 , CH2 and CH3, collectively referred to as CH).
- VL variable domain
- VH variable domain
- CH constant domain
- Fab antigen-binding fragments
- Fc crystallizable fragment
- the constant region domains of the light (CL) and heavy (CH) chains confer important biological properties such as antibody chain association, secretion, trans-placental mobility, complement binding, and binding to Fc receptors.
- the Fv fragment is the N-terminal part of the Fab fragment of an antibody and consists of the variable portions of one light chain and one heavy chain.
- the specificity of the antibody resides in the structural complementarity between the antibody combining site and the antigenic determinant.
- Antibody combining sites are made up of residues that are primarily from the hypervariable or complementarity determining regions (CDRs). Occasionally, residues from non-hypervariable or framework regions (FR, meaning amino acid sequences interposed between CDRs) influence the overall domain structure.
- CDRs refer to amino acid sequences which together define the binding affinity and specificity of the natural Fv region of a native antibody binding site.
- the light and heavy chains of an antibody each have three CDRs, designated L-CDR1 , L-CDR2, L- CDR3 and H-CDR1 , H- CDR2, H-CDR3, respectively.
- An antigen-binding site therefore, includes six CDRs, comprising the CDR set from each of a heavy and a light chain variable region.
- antibody fragment refers to at least one portion of an intact antibody, preferably the antigen binding region or variable region of the intact antibody, that retains the ability to specifically interact with (e.g., by binding, steric hindrance, stabilizing/destabilizing, spatial distribution) an epitope of an antigen.
- antibody fragments include, but are not limited to, Fab, Fab', F(ab')2, Fv fragments, single chain antibody molecules, in particular scFv (single chain variable fragments), disulfide-l inked Fvs (sdFv), a Fd fragment consisting of the VH and CH1 domains, linear antibodies, single domain antibodies such as, for example, sdAb (either VL or VH), camelid VHH domains, multi-specific antibodies formed from antibody fragments such as, for example, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region, and an isolated CDR or other epitope binding fragments of an antibody.
- An antigen binding fragment can also be incorporated into single domain antibodies, maxibodies, minibodies, nanobodies, intrabodies, diabodies, triabodies, tetrabodies, v-NAR and bis-scFv.
- Antigen binding fragments can also be grafted into scaffolds based on polypeptides such as a fibronectin type III.
- the antibody construct of the invention may be a Fab fragment or a single chain antibody.
- the antibody construct is a monoclonal antibody.
- monoclonal antibody refers to a preparation of antibody molecules of single molecular composition.
- a monoclonal antibody refers to a population of antibody molecules that contain only one species of antibody combining site capable of immunoreacting with a particular epitope.
- a monoclonal antibody thus typically displays a single binding affinity for any epitope with which it immunoreacts.
- a monoclonal antibody may therefore contain an antibody molecule having a plurality of antibody combining sites, each immunospecific for a different epitope, e.g., a bispecific monoclonal antibody.
- a monoclonal antibody was produced by immortalization of a clonally pure immunoglobulin secreting cell line, a monoclonally pure population of antibody molecules can also be prepared by other methods known in the art.
- a heavy chain construct or a light chain construct of the invention is able to form an antibody construct of the invention, together with the respective other chain.
- the exact structure of the heavy and light chain constructs thus depends on the desired structure of the antibody construct, e.g., as detailed herein.
- the heavy chain construct and the light chain construct can also be part of the same amino acid chain, e.g., in a single chain antibody such as an scFv.
- a neutralizing antibody or “broadly neutralizing antibody” is one that can neutralize the ability of that pathogen to initiate and/or perpetuate an infection in a host.
- the antibodies produced in accordance with the present invention have neutralizing activity, where the antibody can neutralize at a concentration of 10’ 8 M or lower (e.g., 10’ 9 M, 10’ 10 M, 10’ 11 M, 10’ 12 M or lower).
- binding refers to a direct association between two molecules, due to, for example, covalent, electrostatic, hydrophobic, and ionic and/or hydrogen-bond interactions, including interactions such as salt bridges and water bridges.
- the antibody construct of the present invention has specificity for the ORF2i protein.
- specificity refers to the ability of an antibody to detectably bind to a conformational epitope presented on the ORF2i protein, while having relatively little detectable reactivity with the ORF2g protein and the ORF2c protein. Specificity can be relatively determined by binding or competitive binding assays as known from the art. Specificity can be exhibited by, e.g., an about 10: 1 , about 20: 1 , about 50: 1 , about 100: 1 , 10.000: 1 or greater ratio of affinity/avidity in binding to the specific antigen versus nonspecific binding to other irrelevant molecules.
- affinity means the strength of the binding of an antibody to an epitope. Binding affinity can be described by an antibody's equilibrium dissociation constant (KD), which is defined as the ratio Kd/Ka at equilibrium. Ka is the antibody's association rate constant and Kd is the antibody's dissociation rate constant. Binding affinity is determined by both the association and the dissociation and alone neither high association nor low dissociation can ensure high affinity.
- Ka equilibrium dissociation constant
- Kon on-rate constant
- the association rate constant is expressed in M -1 s -1 , and is symbolized as follows: [Ab]x[Ag]xKon.
- the dissociation rate constant (Kd), or off-rate constant (Koff) measures the number of dissociation events per unit time propensity of an antibody-antigen complex to separate (dissociate) reversibly into its component molecules, namely the antibody and the antigen.
- the dissociation rate constant is expressed in s -1 , and is symbolized as follows: [Ab+Ag]xKoff.
- the equilibrium dissociation constant measures the rate at which new antibody-antigen complexes formed equals the rate at which antibody-antigen complexes dissociate at equilibrium.
- the antibody construct of the invention has an equilibrium dissociation constant KD for non-glycosylated form of pORF2 of less than 100 nM, less than 50 nM less than 10 nM, less than 5 nM, less than 2 nM, less than 1 nM or preferably less than 0.5 nM.
- epitope refers to a specific arrangement of amino acids located on a protein to which an antibody binds. Epitopes often consist of a chemically active surface grouping of molecules such as amino acids or sugar side chains, and have specific three-dimensional structural characteristics, as well as specific charge characteristics. Epitopes can be linear or conformational, i.e. , involving two or more sequences of amino acids in various regions of the antigen that may not necessarily be contiguous.
- formational epitope refers to amino acid residues that are, at least in part, discontinuous in the epitope protein sequence yet come within close proximity to form an antigenic surface on the protein's three-dimensional structure.
- polypeptide refers to a compound comprised of amino acid residues covalently linked by peptide bonds.
- a polypeptide is not limited to a specific length: it must contain at least two amino acids, and no limitation is placed on the maximum number of amino acids that can comprise a polypeptide's sequence.
- Peptides, oligopeptides, and proteins are included within the definition of polypeptide, and such terms may be used interchangeably herein unless specifically indicated otherwise.
- the term refers to both short chains, which also commonly are referred to in the art as peptides, oligopeptides and oligomers, for example, and to longer chains, which generally are referred to in the art as proteins, of which there are many types.
- the term “peptides” refers to a linear polymer of amino acids linked together by peptide bonds, preferably having a chain length of less than about 50 amino acids residues; a "polypeptide” refers to a linear polymer of at least 50 amino acids linked together by peptide bonds, and a protein specifically refers to a functional entity formed of one or more peptides or polypeptides, optionally of non-polypeptides cofactors.
- polypeptide may be an entire protein, or a subsequence thereof.
- Polypeptides include, for example, biologically active fragments, substantially homologous polypeptides, oligopeptides, homodimers, heterodimers, variants of polypeptides, modified polypeptides, derivatives, analogs, fusion proteins, among others.
- a polypeptide includes a natural peptide, a recombinant peptide, or a combination thereof.
- glycosylated with respect to a protein means that a carbohydrate moiety is present at one or more sites of the protein molecule.
- a glycosylated protein refers to a protein that is typically modified by N-glycan or O-glycan addition.
- ORF2 refers to the open reading frame encoding the HEV ORF2 viral capsid protein (“pORF2”).
- the ORF2 protein sequence contains 660 amino acids, has a signal peptide at its N-terminus (that can shuttle it to the extracellular compartment, depending on the start codon), and is further organized into three domains designated as shell (S, amino acids 129 - 319), middle (M, amino acids 320 - 455), and protruding domain (P, amino acids 456 - 606).
- N-X-S/T Three highly conserved potential N-glycosylation sites represented by the sequon Asn-X-Ser/Thr (N-X-S/T) have been identified in pORF2, but N-linked glycosylation has only been confirmed to occur at positions N137 and N562.
- HEV infection leads to the production of at least 3 forms of pORF2 capsid protein: infectious ORF2 (ORF2i), glycosylated ORF2 (ORF2g), and cleaved ORF2 (ORF2c), as shown by adjoin et al. (2018. “Hepatitis E virus lifecycle and identification of 3 forms of the ORF2 capsid protein” Gastroenterology 154 (1): 211-223).
- the ORF2i protein is the structural component of infectious particles, and it is not glycosylated.
- ORF2g and ORF2c proteins are secreted in large amounts in culture supernatant and sera of infected patients, are sialylated, N- and O-glycosylated but are not associated with infectious virions.
- ORF2g and ORF2c proteins the most abundant antigens detected in patient sera, might inhibit antibody-mediated neutralization.
- the present invention provides antigen-binding proteins, antibody constructs, such as antibodies and antigen-binding fragments thereof, preferably monoclonal antibodies, that specifically bind to a non-glycosylated (infectious) HEV particle, or an antigenic fragment thereof, but not to a glycosylated (non-infectious) HEV particle.
- the present invention provides antibody constructs, such as antibodies and antigen-binding fragments thereof, preferably monoclonal antibodies, that specifically bind to a conformational epitope of non-glycosylated pORF2, preferably that specifically bind to a non-glycosylated conformational epitope at the dimerization interface at the tip of the P domain dimer of pORF2 containing glycosylation site N562.
- antibody constructs such as antibodies and antigen-binding fragments thereof, preferably monoclonal antibodies, that specifically bind to a conformational epitope of non-glycosylated pORF2, preferably that specifically bind to a non-glycosylated conformational epitope at the dimerization interface at the tip of the P domain dimer of pORF2 containing glycosylation site N562.
- said antibody construct e.g., said monoclonal antibody is monoclonal antibody p60.1 against HEV, which can specifically bind and neutralize non-glycosylated (infectious) HEV particles. It can also be a derivative thereof, preferably, sharing the CDRs and, optionally, the variable regions thereof .
- said antibody constructs e.g., said monoclonal antibody is monoclonal antibody p60.12 against HEV, which can specifically bind and neutralize non-glycosylated (infectious) HEV particles. It can also be a derivative thereof, preferably, sharing the CDRs and, optionally, the variable regions thereof .
- the antibody constructs of the invention recognize a quaternary epitope that assembles at the dimerization interface at the tip of the P domain dimer of pORF2.
- the broad neutralizing antibodies of the invention bind predominantly to one protomer, but make contacts also with the other one, resulting in an asymmetric binding mode.
- a correctly folded pORF2 dimer is required for antigen-antibody interaction of said antibodies with HEV according to the invention.
- the antibodies of the invention recognize conformation-sensitive epitopes comprising three segments distant in the amino acid sequence of pORF2: 1) amino acids 480-490, 2) amino acids 555-565 and 3) amino acids 580-590.
- N562 of both protomers which serve as attachment points for the N-linked glycans in the secreted pORF2 dimer, form hydrogen bonds with both antibodies, so that such an attached glycan would preclude binding of these bnAbs via steric clashes and thereby causes glycan sensitivity of the antibodies.
- the CDRs of the heavy chain variable region correspond to the amino acid sequences as set forth in SEQ ID NO:2 (p60.1-HC CDR1), SEQ ID NO:3 (p60.1-HC CDR2) and SEQ ID NO:4 (p60.1 -HC CDR3).
- the CDRs of the light chain variable region correspond to the amino acid sequences as set forth in SEQ ID NO:6 (p60.1-LC CDR1), the sequence AAS (p60.1-LC CDR2) and SEQ ID NO:8 (p60.1-LC CDR3).
- the CDRs of the heavy chain variable region correspond to the amino acid sequences as set forth in SEQ ID NO:10 (p60.12-HC CDR1), SEQ ID NO:11 (p60.12-HC CDR2) and SEQ ID NO:12 (p60.12-HC CDR3).
- the CDRs of the light chain variable region correspond to the amino acid sequences as set forth in SEQ ID NO: 14 (p60.1-LC CDR1), the sequence DVT (p60.12-LC CDR2) and SEQ ID NO: 16 (p60.12-LC CDR3).
- the antibodies of the invention may be human antibodies.
- human antibodies includes antibodies having variable and constant regions derived from human germline immunoglobulin sequences whether in a human cell or grafted into a non-human cell, e.g., a mouse cell.
- the human antibodies of the invention 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).
- the term includes antibodies recombinantly produced in a non-human mammal or in cells of a non-human mammal.
- human antibody is not intended to include antibodies in which CDR sequences derived from the germline of another mammalian species, such as a mouse, have been grafted onto human framework sequences (e.g., humanized antibodies).
- the antibodies may alternatively be non-human antibodies, e.g., murinized antibodies or rat antibodies, e.g., for in vivo experiments in mice or rats.
- the present invention also relates to conserved variants or active fragments of any one of the antibody constructs of the invention sharing the CDRs of p60.1 or p60.12, wherein one or more of amino acid residues in said variant or active fragment thereof have been conservatively substituted, added or deleted, wherein the variant or fragment still retains the capability of specifically binding to a conformational epitope of a non-glycosylated polypeptide encoded by hepatitis E virus ORF2.
- conserved variants used herein means that the variants substantially retain the parent's properties, such as basic immunological properties, structural properties, regulating properties or biochemical properties.
- amino acid sequence of the conserved variants of the polypeptide is limitedly different from the parent polypeptide such that the conserved variants and the parent polypeptide are closely similar as a whole and are identical in a lot of regions.
- the difference of amino acid sequence between the conserved variants and parent polypeptide can be replacement, addition and deletion of one or more amino acid residues or any combination thereof.
- the replaced or added amino acid residues may or may not be encoded by genetic code.
- the conserved variants of the polypeptide may be variants produced spontaneously or not spontaneously.
- polypeptide's conserved variants produced not spontaneously may be produced by induced mutation techniques or by direct synthesis. According to the disclosed content of the invention, a person skilled in the art would appreciate that the fragment of said antibody of the invention may be modified to substantially preserve the property of specifically binding to hepatitis E virus of the generated antibody variants.
- any deviations from the defined SEQ ID NO: may be conservative substitutions, wherein, preferably, there is at most one conservative substitution per chain.
- Conservative substitutions are known in the art, wherein an amino acid is exchanged for a different amino acid with similar biochemical properties, e.g., charge, hydrophobicity and/or size.
- substitutions are selected from the same class of amino acids, as defined herein:
- the sequence identity in the CDR1 , CDR2, and/or CDR3 may be 100% (to the respective defined CDRs).
- sequence identity in all CDRs or a heavy chain consruct and/or a light chain construct is 100%.
- sequence identity in the variable regions also is 100%, i.e., the variable regions of the antibody construct or the inventions correspond to the variable regions of p60.1 or p60.2.
- the invention does not only provide the heavy and light chain constructs, but also an antibody construct comprising the heavy chain construct and light chain construct of the invention, i.e., an antibody construct capable of specifically binding to a conformational epitope of a non-glycosylated polypeptide encoded by hepatitis E virus ORF2.
- an antibody construct capable of specifically binding to a conformational epitope of a non-glycosylated polypeptide encoded by hepatitis E virus ORF2.
- said antibody constructs have been found to be particularly advantageous for diagnostic, therapeutic and prophylactic uses.
- nucleic acid molecule encoding a heavy chain construct and/or light chain construct of the invention or an antibody construct of the invention.
- a is understood to mean "at least one", i.e., a nucleic acid molecule of the invention may also encode a heavy chain construct of the invention and a light chain construct of the invention, and preferably encodes an antibody constructs of the invention.
- Said nucleic acid molecule may be a recombinant expression vector comprising said nucleic acid molecule, preferably, under the control of a heterologous promoter.
- the promotor preferably is able to mediate expression in a host cell, e.g., a bacterial cell, a yeast cell, a eukaryotic cell, an insect cell, a mammalian cell such as a CHO cell or a human cell, e.g., a B cell, a T cell or an NK cell.
- a host cell e.g., a bacterial cell, a yeast cell, a eukaryotic cell, an insect cell, a mammalian cell such as a CHO cell or a human cell, e.g., a B cell, a T cell or an NK cell.
- the promotor can be a constitutive promotor or an inducible promotor.
- the skilled in the art can get the nucleotide sequences encoding the same and get their variants according to codon degeneracy.
- the nucleic acid is codon-optimized for expression in the desired host cell.
- the skilled in the art can get recombinant expression vectors comprising the nucleotide sequences described above, and host cells transformed with the expression vectors by many methods. Selection of host cells and transformation technologies that can be used to express said antibodies or their active fragments are well known from the art.
- the present invention also relates to a recombinant expression vector comprising the nucleic acid molecule encoding said antibodies or active fragments.
- vector any genetic element, such as a plasmid, minicircle, YAC, phage, transposon, e.g., suitable for being transposed by Sleeping Beauty, cosmid, chromosome, a viral vector or virus, e.g., a retroviral or adenoviral vector etc., which is capable of replication when associated with the proper control elements and which can transfer gene sequences between cells.
- the term includes cloning and expression vectors, as well as viral vectors.
- the antibodies of the invention and/or active fragments thereof can be expressed in proper host cells by genetic engineering methods known in the art.
- the invention further relates to host cells transformed with said recombinant expression vectors.
- Many expression host cells can be used in the present invention, for example, prokaryotic cells including but not limited to Escherichia coli, Bacillus, Streptomyces, eukaryotic cells including but not limited to Aspergillus, Saccharomycetes, as well as mammalian cells, plant cells and so on.
- the expression of the antibody constructs of the invention is not limited to any specific expression vectors or host cells, as long as they can be used to express said antibodies.
- the invention thus also provides a host cell comprising said recombinant expression vector, which is able to express the heavy chain construct and/or light chain construct or the antibody construct of the invention.
- the host cell expresses said construct, e.g., said antibody construct, on its cell surface (e.g., in case of a CAR or a transmembrane immunoglobulin), or it secretes said construct (typically, in the case of an antibody, e.g., a single chain antibody).
- the cell may be a human cell, e.g., a B cell, a T cell or an NK cell.
- the antibody constructs of the present invention bind to antigens present in sera of patients infected with HEV.
- the present invention also relates to said antibody constructs for use in diagnosis of hepatitis E virus infection.
- a further object of the present invention relates to a method for detecting the presence of native, non-glycosylated HEV pORF2 in a sample, e.g., a sample from a patient, comprising contacting the sample with the antibody constructs of the present invention under conditions that allow an immunocomplex of the protein and the antibodies to form, wherein detection of the immunocomplex indicates the presence of native, nonglycosylated HEV pORF2 in the sample (for instance: immunoprecipitation, immunofluorescence or western blotting).
- infectious HEV particles may be detected using the antibody constructs of the present invention because these antibody constructs, e.g., monoclonal antibodies, recognize specific antigenic determinants for infectious HEV particles located on the surface of HEV.
- a further object of the present invention relates to a method for detecting the presence of infectious HEV particles in a sample comprising contacting the sample with the antibodies of the invention under conditions that allow an immunocomplex of the antibody and the infectious particles to form, wherein detection of the immunocomplex indicates the presence of the infectious particles in the sample.
- sample includes any solid or fluid sample, liable to contain infectious particles of hepatitis E virus.
- the sample is selected from the group consisting of ascites, urine, saliva, sweat, milk, synovial fluid, peritoneal fluid, amniotic fluid, percerebrospinal fluid, lymph fluid, lung embolism, cerebrospinal fluid, and pericardial fluid.
- the sample is a feces sample.
- the sample is a urine sample.
- the sample is a saliva sample.
- the sample is a blood sample.
- blood sample means any blood sample derived from a subject.
- the detecting methods of the present invention are particularly suitable for diagnosing acute HEV infection, recent HEV infection, chronic HEV infection, weak active HEV infection or cleared HEV infection.
- Assays and conditions for the detection of immunocomplexes are known to those of skill in the art. Such assays include, for example, competition assays, direct reaction assays sandwich-type assays and immunoassays (e.g., ELISA).
- the assays may be quantitative or qualitative. There are a number of different conventional assays for detecting formation of an antibody-peptide complex comprising an antibody of the present invention.
- the detecting step can comprise performing an ELISA assay, performing a lateral flow immunoassay, performing an agglutination assay, analyzing the sample in an analytical rotor, or analyzing the sample with an electrochemical, optical, oropto-electronic sensor.
- the antibody constructs of the present invention may be used in highly sensitive methods for screening and identifying individuals carrying HEV and/or infected with HEV, as well as for screening for HEV-contaminated samples.
- the antibody constructs of the present invention may also be used in assays for monitoring the progress of anti-HEV therapies in treated individuals, or for monitoring the growth rate of HEV cultures used in research and investigation of the HEV agent.
- kits for the diagnosis of hepatitis E virus infection comprising at least one of the antibody constructs of the invention, or any combination thereof (e.g., an antibody construct based on p60.1 and an antibody construct based on p60.12).
- the kit may comprise the antibody construct in an amount effective for diagnosis. It optionally comprises detection agents suitable for detection of the interaction of antigen and antibody construct corresponding to the adopted detection method.
- the kit may also comprise suitable buffers.
- the term "amount effective for diagnosis” is intended to mean the antibody construct of the invention, conserved variants or active fragments thereof at the amount effective for detection of HEV in a biological sample.
- the amount of the above-mentioned material would be variable depending on the different immunochemical methods used.
- the diagnostic kit where appropriate, should further include suitable absorbent carriers, buffer reagent/solutions, reagents used to produce visible signals fortesting and instructions for use. Examples of kits include but are not limited to ELISA assay kits, and kits comprising test strips and dipsticks.
- the antibody constructs of the invention specifically recognize antigenic determinants located on the surface of infectious HEV particles, showing high binding affinity, potent neutralization activity, as well as broad reactivity across HEV genotypes.
- the antibodies of the invention efficiently neutralize naked and quasi-enveloped viral particles. Said antibodies may therefore be used for the prophylaxis and/or for the passive immunization treatment of HEV infections, especially for chronical and immunocompromised HEV patients.
- the invention also provides a pharmaceutical composition
- a pharmaceutical composition comprising a) an antibody construct of the invention, b) an expression vector of the invention, wherein the nucleic acid encodes the antibody construct of the invention and/or c) a host cell of the invention, wherein the nucleic acid encodes the antibody construct of the invention, and, optionally, a pharmaceutically acceptable vehicle and/or excipient.
- Said pharmaceutical composition may be for use in preventing or treating hepatitis E virus infection. They may comprise a pharmaceutically effective amount of said antibody construct.
- the present invention relates to the antibody constructs of the invention for use as a medicament for prophylaxis and/or treatment of HEV infection.
- the pharmaceutical composition of the invention When supplied prophylactically , the pharmaceutical composition of the invention is provided in advance of any exposure to any one or more of the HEV strains or in advance of any symptoms due to infection of the viruses.
- the prophylactic administration of the pharmaceutical composition of the invention serves to prevent, reduce the risk or likelihood of or attenuate any subsequent infection of these viruses in a mammal, preferably in a human subject.
- the pharmaceutical composition of the invention is provided at or after (preferably, shortly after) the onset of infection or at the onset of any symptom of infection or any disease or deleterious effects caused by these viruses.
- the therapeutic administration of the pharmaceutical composition of the invention serves to attenuate the infection or disease.
- the pharmaceutical composition of the present invention may, thus, be provided either prior to the anticipated exposure to HEV or after the initiation of infection. According to the specific conditions of the subject to be treated, the skilled in the art know how to select proper doses and administration routes.
- the pharmaceutical composition of the invention may also comprise a therapeutically effective amount of the antibodies of the invention combined with at least one other anti-viral agent as an additional active ingredient.
- agents may include but are not limited to interferons, other anti HEV monoclonal antibodies, anti HEV polyclonal antibodies, RNA polymerase inhibitors, protease inhibitors, IRES inhibitors, helicase inhibitors, immunomodulators, antisense compounds and ribozymes.
- a method of treating a subject infected with HEV or of reducing the likelihood of infection of a subject at risk of contracting HEV comprising delivering to said subject a prophylactically effective amount or a therapeutically effective amount of at least one of the antibody constructs of the invention.
- prophylactically effective amount can be replaced with the term “immunologically effective amount", which means the amount sufficient to elicit immune prevention for a subject. It is well known that the “prophylactically effective amount” may vary according for example to the administration way, the characteristics of the individual subject, and the antibody construct used. According to the published references, teachings and corresponding clinical criteria in the art, the “prophylactically effective amount” of an antibody construct can be determined.
- the preferable prophylactically I immunologically effective amount is 0.0001 mg - 0.1 mg per dose.
- the term "therapeutically effective amount” means the amount sufficient to elicit effective protection for the subjects and to neutralize HEV, or an amount effective in alleviating the symptoms of the HEV infection or reducing the number of circulating viral particles in an individual. It is well known that a therapeutically effective amount of an antibody may vary depending on different treatment schemes, illness courses, characteristics of the individual subject, and the antibodies used. According to the published references, teachings and corresponding clinical criteria in the art, a clinician can determine the "therapeutically effective amount" of an antibody construct based on their own experience. The preferable therapeutically effective amount is 0.001 mg - 20 mg per kg weight.
- the antibody constructs of the invention based on p60.1 are particularly effective in neutralizing HEV g3, g4 and g2, and the constructs based on p60.12 are particularly effective in neutralizing HEV g3, g4 and rat-HEV, they are preferably used for diagnosis, treatment or prevention of said virus subtypes.
- the P domain serves as the receptor-binding domain of HEV and is thus a primary target for neutralizing antibodies.
- pORF2 exhibits both secreted and non-secreted forms during infections. The former are present as glycosylated dimers in sera of infected patients, while the later are components of the infectious particle.
- constructs for all genotypes were designed to represent the non-secreted form of the P domain.
- a construct representing the secreted form was designed using HEV genotype 3 P domain (Table 1).
- the pGS plasmids according to Table 1 verified by sequencing to carry the desired inserts were transfected into Drosophila S2 cells essentially as described previously (Johansson et al. 2012. Production of Recombinant Antibodies in Drosophila mela nogaster S2 Cells. Patrick Chames (ed.), Antibody Engineering: Methods and Protocols, Second Edition, Methods in Molecular Biology, 907). Briefly, stable cell lines were generated by cotransfecting the plasmids coding for the gene of interest with a puromycin-resistance selection plasmid. Following selection with puromycin and adaptation of the cells to insect- Xpress media, cells were induced with CdCh for large-scale protein production. On day five post-induction, the intracellular and the secreted P domains were affinity purified from cell lysates and culture supernatants, respectively. Protein purity assessment was performed by SDS-PAGE analysis.
- antigen-specific memory B cells were isolated from peripheral blood mononuclear cells (PBMCs) from two convalescent HEV patients (N1960 (p60), 4100869889, 8.5 x 10 7 cells and N1961 (p61), 1707407510, 1.44 x 10 8 cells) by magnetic activated cell sorting (MACs). MACs-sorted cells were then stained with fluorescently labeled P domains and analyzed for the frequency and phenotype of antigen-specific B cells by flow cytometry prior to sorting of antigen-specific B cells. This was then followed by single B cell RNA sequencing as described in the next sections.
- PBMCs peripheral blood mononuclear cells
- MACs-sorted cells were then stained with fluorescently labeled P domains and analyzed for the frequency and phenotype of antigen-specific B cells by flow cytometry prior to sorting of antigen-specific B cells. This was then followed by single B cell RNA sequencing as described in the next sections.
- PBMCs from two convalescent HEV patients were isolated using density gradient separation medium (Histopaque, Sigma Aldrich) according to the manufacturer’s instructions and stored at -150 °C in 10% DMSO and 90% (v/v) fetal bovine serum.
- B cells were obtained from PBMCs by magnetic separation using CD19 microbeads and stained with anti-human CD20-Alexa Fluor 700, anti-human IgG-APC, LIVE/DEADTM Fixable Near-IR Dead Cell Stain, and recombinant differentially fluorescently labelled HEV gt3 P domains (pGS38 and pGS39) with the aim of obtaining antibodies that bind the nonglycosylated form but not the glycosylated form.
- Approximately 4,170 and 19,760 live cells were sorted for p60 and p61 , respectively, using band pass filters R 780/60, R 730/45, R 670/30, YG 610/20 and B 530/30.
- Antigen-specific memory B cells were single-cell sorted using 10xGenomics technology and B cell receptor sequences obtained by Next Generation Sequencing using Illumina technology. The obtained FASTQ files were used to generate single cell V(D)J sequences and annotations using Cell ranger vdj (version 3.1.0). Output files were loaded and analyzed with the Loupe V(D)J Browser (10x Genomics).
- variable regions of antibodies were selected after analysis of the sequences using the Loupe V(D)J browser based on a criterion described in the result section below.
- the variable region of the heavy chain was covalently linked via its C-terminus to the N-terminus of the respective light chain variable region using a 3x glycine-serine linker (GS-linker).
- Codon-optimized synthetic genes of the covalently linked paired sequences cloned into a pMT vector were purchased from Twist biosciences. All constructs carried a Drosophila BiP signal sequence at the N-terminus as well as an EK cleavage site and a double strep-tag at the C-terminus.
- the name of the individual scFvs was defined by scFv as a prefix, followed by the donor ID and the clone number. Expression and purification of all scFvs followed the protocols in sections 1.1.3.
- the P domains and TEV protease were mixed in a 1 :4 ratio and digestion done in SEC buffer at 28 °C for 4 hours.
- a reaction mix for EK digestion was set up containing 2 mg protein, 300pl 10x EK buffer (500 mM NaCI, 20 mM CaCh, 200 mM Tris pH 8), and 100 pl EKMaxTM enterokinase (Thermo Scientific) diluted 1 :25 in 1x EK buffer. This was then topped up to 3 ml with Milli-Q water and incubated at 28 °C overnight, after which 30 pl 100 mM PMSF was added to inactivate the enterokinase. Since crystallization requires large amounts of protein, the quantity of protein digested was adjusted accordingly.
- the digestion product was flown through a strep-tactin column, thereby retaining the undigested protein still carrying the strep tag.
- the digestion product was applied onto a 1 ml HisTrapTM excel column (Cytiva), thereby retaining the TEV in the column.
- the flow-through and wash fractions that contained the digested proteins were pooled and further purified by size exclusion chromatography.
- the elution fractions containing the proteins of interest were pooled, concentrated, and stored at -80°C.
- Crystallization experiments of HEV gt3 P domain in complex with the best neutralizing scFvs involved mixing pGS99 with scFvs (1 :1.2 molar ratio, respectively) and incubating the mixture overnight at 16 °C.
- the complexes were purified on a superdex 200 increase 10/300 GL column (Cytiva) equilibrated in gel filtration buffer.
- the purified complexes were then concentrated to 9.3 mg/ml (scFv-p60.1-pGS99) and 6.7 mg/ml (Fab-p60.12-pGS99).
- Paired heavy and light chain variable regions of antibodies were amplified from the respective top neutralizing scFvs. These were inserted into a pcDNA3.1 expression vector.
- the heavy chain variable regions were cloned into the IgG 1 expression vector, while their respective kappa or lambda pairs were cloned into expression vectors carrying the corresponding constant regions. All genes were cloned downstream a CD5 signal peptide to allow the secretion of mature IgGs. Plasmid DNA was amplified, isolated and sequenced.
- HEK Expi293FTM cell lines were thawed and maintained in Expi293TM expression medium (Gibco) according to the supplier’s manual and cultured at 37 °C, 8 % CO2, with shaking at 125 rpm.
- the ExpiFectamineTM 293 transfection kit (Gibco) was used with slight modifications. A day before transfection, 2.5 x 10 6 viable cells/ml were seeded and cultured overnight in a 500 ml Erlenmeyer flask. Cell density and viability were determined by trypan blue exclusion on a Countess II FL cell counter (Life technologies).
- the required volume of cells was centrifuged for 5 minutes at 300x g before carefully resuspending the cells in fresh media.
- the overnight culture with a viability > 90 % was diluted to 2 x 10 6 viable cells/ml in 85 ml pre-warmed medium, and cells were returned to the incubator.
- the plasmid DNA cocktail was prepared in 5 ml Opti-MEM (Gibco) at ratios listed in Table 3, resulting in total plasmid DNA of 1 pg per ml of culture volume transfected.
- addition of p21 , p27 and SV40 serves to enhance protein expression in HEK cells.
- the ExpiFectamineTM 293 transfection reagent was then prepared by mixing 266.7 pl of the reagent with 4733.3 pl Opti-MEMTM and allowed to incubate for 5 minutes at room temperature. Following the 5-minute incubation, the diluted reagent was mixed with the plasmid DNA cocktail by inversion. The ExpiFectamineTM 293/DNA complexes were incubated for 20 minutes at room temperature, after which they were added to the cells dropwise, and the cells were subsequently incubated for 16-22 hours. This was followed by dropwise addition of 500 pl ExpiFectamineTM 293 transfection Enhancer 1 and 5 ml ExpiFectamineTM 293 transfection Enhancer 2 to the transfected cells, and the cells were returned to the incubator.
- Plasmids Amount (pq/ml of transfection) pq/100 ml transfection volume
- the cells were harvested by adding 10 ml 10x PBS and centrifugation at 70,000x g for 20 minutes. The supernatant was filtered through a 0.22 pm syringe filter prior to loading onto a 1 ml HiTrap Protein-G column (Cytiva) connected to the AKTATM pure 25 system. Unbound proteins were washed off with 1x PBS and IgGs eluted in 0.1 M Glycine pH 2.7, which was then neutralized by adding 100 pl 1 M Tris-HCI pH 8. The eluted neutralized IgGs were buffer exchanged to PBS using a superdex 200 increase 10/300 GL column. The elution fractions containing the protein of interest were pooled, concentrated and stored at 4 °C until needed for further experiments (neutralization assays, ELISA and Biacore).
- Strep-TactinXT (I BA Life sciences) was amine coupled to a carboxyl-derivatized CM-5 sensor chip (Cytiva) using a Biacore 3000 (GE Healthcare) following the protocol from the Twin-Strep-tag capture kit for surface plasmon resonance (I BA Life sciences).
- the chip surfaces were prepared by pre-treatment with three consecutive 1 -minute pulse injections of 50 mM NaOH. The surfaces were then activated for 10 minutes with a 1 :1 mixture of NHS and EDC at a flow rate of 10 pl/min. 35 pg/ml Strep-TactinXT in 10 mM sodium acetate pH 4.5 was immobilized for 10 minutes at a density of 3922 RU on all flow cells.
- HBS-EP buffer 25 nM ligand (26 kDa, HEV gt3 secreted and non-secreted P domains) in HBS-EP buffer was injected for 1 minute over flow cell 2 at a flow rate of 10 pl/min. Then, the analytes (lgG-p60.1 and p60.12) in HBS-EP buffer were injected over two flow cells (1 and 2) at concentrations of 75, 50, 25, 10 and 5 nM at a flow rate of 30 pl/min and a temperature of 25 °C. The complex was allowed to associate and dissociate for 3.33 and 7 minutes, respectively. The surfaces were then regenerated with a 45-second injection of 3 M guanidine hydrochloride. Measurements were performed twice per sample, and the individual curves were used to produce the mean affinity constant by global fitting to a 1 :1 binding model using the BiaEvaluation software.
- HEV gt3 P domain comprising amino acid 456-660 (UniProtKB accession number: C4B4T9; SEQ ID NO: 17) fused to fluorescent proteins
- the constructs carried either mNeonGreen or mRuby at the N-terminus for the non-secreted (pGS38) and secreted form (pGS39), respectively ( Figure 1 B).
- the secreted form was cloned into a pMT vector carrying a BiP signal sequence while the non-secreted form was inserted into the same vector without the signal peptide for expression in Drosophila S2 cells as described above.
- a stable S2 transfectant was established per construct and the proteins produced as described above.
- the retention volume of both proteins was approximately 190 ml on a HiLoad 26/600 superdex 200 pg (Cytiva), but the profile of the secreted P domain showed an additional peak eluting slightly earlier. Since the P domains form dimers, the elution volume was expected, but the additional peak observed for the secreted P domain suggested a tendency to form higher oligomers.
- Both intracellular and secreted proteins showed > 90 % purity as judged from analysis by SDS-PAGE using a 12 % gel under reducing conditions followed by Coomassie staining. Interestingly, the apparent molecular weight of the secreted P domain was slightly higher than the intracellular protein, a difference that can be attributed to the attached glycan at position 562 in the secreted construct.
- sequence pairs 50 from donor p60 and 41 from donor p61 , were selected for expression in Drosophila S2 cells.
- the sequences represented 6 germline genes, and varied in heavy-chain complementarity determining region-3 (CDRH3) amino acid lengths (8-25 aa long), according to IMGT numbering, as well as somatic hypermutations (2-37 nucleotide substitutions per VH sequence).
- CDRH3 heavy-chain complementarity determining region-3
- Synthetic genes of the selected 91 -paired sequences, cloned into a pMT vector as singlechain fragments of the variable region (scFv) were purchased from Twist biosciences. The name was defined by scFv as a prefix, followed by the donor ID and the clone number. All scFvs carried a C-terminal EK site and a double strep-tag to aid purification. Furthermore, they were cloned downstream a BiP signal sequence to allow expression through the secretory pathway. A stable S2 cell line was generated per scFv as described above, and a similar protocol was followed for expression and purification of the scFvs.
- HEV gt3 Kemow-C1 p6 clone produced as previously described (Todt et al. 2020. Robust hepatitis E virus infection and transcriptional response in human hepatocytes PNAS 117 (3): 1731-1741) were performed.
- HepG2 cells were transfected with the viral genome by electroporation. Four days after transfection, virus was harvested. To generate viral stocks containing naked viral particles, transfected cells were lysed by three freeze-thaw cycles using liquid nitrogen. For virus stocks containing pseudo-enveloped particles, the transfected cells supernatants were used after filtration (0.45pm).
- Indicated virus stocks were incubated with different mAb concentrations for 1 hour at room temperature before infecting HepG2/C3a Hepatoma cell lines. Cells were then incubated for 24 hours before the addition of new media. After four days, cells were fixed with paraformaldehyde and treated with 0.2 % Triton X-100 solution for 5 minutes at room temperature for permeabilization. Subsequently, samples were washed and stained overnight with an anti-ORF2 rabbit polyclonal antibody. Infected cells were visualized following staining with Alexa Fluor 488-labeled goat anti-rabbit antibody, and focus forming units (FFU) counted using the Elispot CTL system (Immunospot).
- FFU focus forming units
- IC50 half-maximal inhibitory concentration
- Figures 2 C and D show neutralization results with pseudo-enveloped viral particles (IC50 values for p60.1 and p60.12 are 0.27 pg/ml and 0.12 pg/ml).
- IC50 values for p60.1 and p60.12 are 0.27 pg/ml and 0.12 pg/ml.
- Neutralization of naked viral particles of another genotype 3 viral strain (HEV83-2-27) at a concentration of 10pg/ml is shown in figure 2 E.
- mAb p60.1 showed neutralization of more than 80% whereas p60.12 neutralized more than 50% of the virus.
- variable region genes for 15 scFvs with IC50 ⁇ 0.5pg/ml were cloned into plasmids for expression of human lgG1 , and expressed in Expi293F cells. All purified antibodies eluted as homogenous single heterodimeric peaks ( Figure 3 A) with > 95 % purity upon analysis on a Coomassie-stained reducing SDS-PAGE gel ( Figure 3 B).
- genes encoding the P domains (amino acid residues 456-660) from the four human pathogenic genotypes and a rat-HEV isolate (Table 2) were cloned into a pMT vector for expression in Drosophila S2 cells.
- the proteins carried both a TEV and EK protease cleavage sites in tandem, in addition to an N-terminal double strep-tag.
- the intracellular form of the P domains was used except in the case of genotype 3.
- both the secreted and intracellular forms were cloned.
- Stably transfected S2 cells were generated, protein expression induced, and purified as described above.
- the retention volume for the dimers was approximately 200 ml, however, the rat intracellular (pGS135) and genotype 3 secreted (pGS100) P domains showed a similar tendency to form higher oligomers. Nevertheless, all proteins showed high purity upon analysis on a Coomassie- stained 15 % SDS-PAGE gel loaded under reducing conditions.
- the HEV gt3 secreted P domain (pGS100) migrated slightly slower than the intracellular domains. Fractions considered in this study were obtained from the dimeric peak.
- Human pathogenic HEV genotypes have a single serotype and the level of amino acid sequence variation ranges between 89 and 93 % across the four genotypes, suggesting high level of conservation.
- the amino acid conservation between the rat-HEV P domain and the human genotypes reaches approximately 31.5 %.
- the level of conservation suggests that it is possible to isolate cross-binding and potentially cross-neutralizing mAbs. Therefore, to explore the cross-genotype binding activity of the mAbs, the purified P domains from section 2.5.2 were used in an indirect ELISA assay as described above.
- the antibodies were tested for their ability to distinguish between the secreted and intracellular P domains.
- the goal was to identify antibodies that targeted only the non-secreted form, a component of the infectious particle, but not a secreted soluble pORF2 dimer that is present in the serum at high levels.
- an indirect ELISA was set-up using HEV gt3 secreted (pGS100) and non-secreted (pGS99) P domains as antigens.
- a sandwich ELISA confirmed binding of both mAb p60.1 and p60.12 to antigens present in sera of three different patients, chronically infected with HEV (Figure 6). All positive samples were highly HEV-RNA positive (between 1x10 5 and 9x10 6 lU/ml) as determined by the central laboratory of Hannover Medical School. Presence of HEV antigens was confirmed by using the commercial WANTAI HEV Ag ELISA. As control, a negative serum as determined by HEV RNA PCR and WANTAI HEV Ag ELISA was used.
- SEC size exclusion chromatography
- the two mAbs were considered for further analysis in SEC because of their IC50 0.01 g/ml.
- the antigen was incubated overnight at 16 °C with a 1 :3 molar ratio (pGS100: scFv) of the respective scFvs. The mixture was centrifuged at maximum speed in a tabletop centrifuge (Eppendorf) for 10 min at 4 °C to remove any precipitate.
- the antibodies were further characterized by surface plasmon resonance (SPR) using a Biacore 3000 (GE Healthcare). Experiments were performed at 25 °C using a Strep- TactinXT CM-5 chip prepared as described above. Briefly, the ligand (HEV gt3 P domains, pGS99 or pGS100) was injected over one flow cell followed by injecting different concentrations of the analytes (mAbs p60.1 , p60.12) over two flow cells. Measurements were fitted to a 1 :1 binding model using the BiaEvaluation software resulting in the sensorgrams of Figure 8 A and B. SPR data showed that the binding of the antibodies towards the intracellular P domain was in the picomolar range, with p60.1 showing the highest affinity binding (Table 5).
- SPR surface plasmon resonance
- association (Ka) and dissociation (Kd) rates for all antibodies using the non-secreted P domain (pGS99) were similar, but the absolute binding (as judged from the number of response units) obtained for p60.1 and p60.12 were lower than for the other antibodies.
- the kinetic parameters for p60.1 and p60.12 using the secreted P domain (pGS100) were not calculated because data was below the threshold recommended in the kit manual (I BA Lifesciences twin-strep-Tag capture kit). Overall, the data show that the antibodies of the invention isolated from HEV-infected patients interact with high affinity with the HEV P domains.
- Protein crystallization was carried out using pGS99 in complex with scFvs or Fabs derived from the mAbs of the invention in order to structurally characterize the antibody-antigen interaction.
- strep-tags Prior to crystallization, strep-tags were proteolytically removed from pGS99 and the scFvs/Fabs using TEV protease and enterokinase, respectively. Thereafter, complex formation was performed by mixing pGS99 and the antibodies (scFv p60.1 and Fab p60.12) in 1 :1.2 molar ratio, and incubating at 16°C overnight. Purification of the complexes by gel filtration showed that the antibody fragments formed complexes with pGS99 ( Figure 9, black arrows), with excess of the antibody fragments observed in both complexes ( Figure 9).
- Crystallization drops were set-up using the purified complexes described above. Crystals for complexes with scFv-p60.1 and Fab-p60.12 were obtained at 293K using sitting drop vapor diffusion method (Figure 10). The conditions that produced diffraction quality crystals per complex are presented in Table 6. The crystals of the complexes were obtained from initial screening conditions and required no further optimization. For data collection, all crystals were flush frozen in mother liquor containing 30 % ethylene glycol. The diffraction capacity of the crystals is listed in Table 6. Table 6: Crystallographic data collection and statistics
- the complex pGS99_scFv-p60.1 crystallized as two P domain dimers each bound by the scFv at the dimer interface per asymmetric unit ( Figure 11). Since the P domain is known to form dimers, the presence of these dimers in the structures of scFv-p60.1 was expected.
- the electron density map of the P domain was well defined and superposition of the complex to the E2s dimer (PDB 3rkc) yielded a root mean square deviation of 0.234 A for all Ca atoms of the E2s domain. This suggests that the conformation of the P domain remains unaltered upon binding of the scFv.
- the linker region connecting the variable heavy and light chains of the scFv was not defined in the electron density map as expected, since this region is known to be intrinsically disordered.
- Both p60.1 and p60.12 recognize a quaternary epitope that assembles at the dimerization interface at the tip of the P domain dimer ( Figure 12).
- Both broad neutralizing antibodies bind predominantly to one protomer, but make contacts also with the other one, resulting in an asymmetric binding mode and indicating that a correctly folded pORF2 dimer is required for antigen-antibody interaction.
- Scrutiny of the interfaces reveals three segments distant in the amino acid sequence that constitute the two epitopes including 1) aa 480- 490, 2) aa 555-565 and 3) aa 580-590, confirming that both antibodies recognize conformation-sensitive epitopes.
- N562 of both protomers which serve as attachment points for the N-linked glycans in the secreted pORF2 dimer, form hydrogen bonds with both antibodies, demonstrating that such an attached glycan would preclude binding of these bnAbs via steric clashes and thereby causing the observed glycan sensitivity.
- scFv-p60.1 In order to gain insight into the biological relevance of the interactions observed in the complexes of the present study, the crystal structure of scFv-p60.1 was superposed onto HEV VLP (PDB 2ztn) ( Figure 13). The superpositions shows that scFv-p60.1 occupies the apex of the dimer. Thus, scFv-p60.1 may act by blocking attachment to the receptor, since its epitope is located in the region proposed to contain the receptor binding site (Mori and Matsuura, 2011. Structure of hepatitis E viral particle. Virus Research 161 (1): 59-64; Xu et al., 2016. Role of asparagine at position 562 in dimerization and immunogenicity of the hepatitis E virus capsid protein. Infection, Genetics and Evolution 37: 99-107).
- HEV gt3 P domains fused to fluorescent proteins for antigen-specific single B cell sorting and sequencing sequences of roughly 3800 antibodies from two HEV convalescent patients were obtained. All the most efficient antibodies in the neutralization assay except one were obtained from patient p60 suggesting differences in response to infection. The variation between patients is not unusual as a number of factors such as age, sex, and comorbidities are known to influence response to infection and disease.
- the isolated antibodies were highly potent in the neutralization assays with HEV gt3, blocking viral infection at concentrations as low as ⁇ 0.01 pg/ml and therefore provide a potential option for treatment of HEV infection.
- the non-secreted form is the component of the infectious particle (Ankavay et al., 2019. New insights into the ORF2 capsid protein, a key player of the hepatitis E virus lifecycle. Scientific Reports, 9(1)), whereas the secreted form is a soluble dimer that is present in the serum at high levels even after infection is cleared (Behrendt etal., 2016. Hepatitis e Virus (HEV) ORF2 Antigen Levels Differentiate between Acute and Chronic HEV Infection. Journal of Infectious Diseases 214 (3): 361-368).
- HEV Hepatitis e Virus
- the secreted form is glycosylated at the conserved glycan position N562 (Xu et al., 2016. Role of asparagine at position 562 in dimerization and immunogenicity of the hepatitis E virus capsid protein. Infection, Genetics and Evolution 37: 99-107; Ankavay et al., 2019). This position is located at the apex of the P domain, which is the receptor-binding region and a binding site for some neutralizing antibodies (Guu et al., 2009. Structure of the hepatitis E virus-like particle suggests mechanisms for virus assembly and receptor binding. PNAS 106 (31): 12992-12997; Mori and Matsuura, 2011. Structure of hepatitis E viral particle.
- mAb 8G12 is a mouse derived cross-genotype antibody against HEV (Gu et al., 2015. Structural basis for the neutralization of hepatitis E virus by a cross-genotype antibody. Cell Research 25 (5): 604-620) that binds at the dimer interface. Residues T563 and T564 are among the contact residues of 8G12 and are located next to N562.
- the close proximity of the glycan therefore may sterically block the binding of 8G12.
- structural studies of the best neutralizing antibodies of the present study revealed that p60.1 and p60.12 bind at the apex of the P domain, directly making contact with the two-asparagine sidechain that serve as attachment site for N-linked glycans at the dimer interface.
- p60.1 makes contact with residues T489, N560, Y561 , N562, T564, T585, and T586 which have been implicated in HEV receptor-interaction (Mori and Matsuura, 2011. Structure of hepatitis E viral particle. In Virus Research 161 (1):59-64.
- C1-C6 Six antigenic cluster sites (C1-C6) targeted by anti-HEV antibodies have been mapped on the P domain by alanine scanning and antibody competition assays (Zhao et al., 2015; Wen et al., 2020).
- a comparison of the epitopes targeted by p60.1 and p60.12 shows that both antibodies occupy antigenic site C3, which is located at the apex of the P domain and is the cell attachment region.
- C2 and C6 are described as the target for the majority of the neutralizing mAbs, with the C6- directed mAbs being the most potent.
- C6 is associated with high neutralization potency and limited or no cross-genotype reactivity, whereas clusters C5 and C3 demonstrate weak or no neutralization potential. This is in contrast to the present invention in which it is shown that surprisingly antibodies targeting antigenic cluster 03, p60.1 and p60.12, are the most potent neutralizers. p60.1 and p60.12 are highly potent neutralizers, demonstrate cross-genotype binding potential and cross-neutralization and may act via blocking virusreceptor interactions and a direct binding competition with the receptor.
- potent neutralizing anti-HEV antibodies from HEV convalescent patients have been isolated. These antibodies bind with picomolar affinities to HEV gt3 P domain, show potent neutralization activity in in vitro assays, and demonstrate broad reactivity across HEV genotypes. The most potent antibodies target only the non-secreted form of the P domain, which is the component of the infectious particle. So far, it is hypothesized that the antibodies of the present study likely act by blocking receptor interaction. Currently, the best antibodies are being tested in an HEV infection model to evaluate their potency in vivo.
- HEV circulates as quasi-enveloped virions in the blood and such virions have been so far reported to be resistant to antibody neutralization (Yin et al., 2016. Distinct Entry Mechanisms for Nonenveloped and Quasi-Enveloped Hepatitis E Viruses. Journal of Virology 90 (8): 4232-4242).
- the antibodies isolated in the present invention efficiently neutralize also quasi-enveloped virions and therefore demonstrate their potential therapeutic benefits for chronical and immunocompromised HEV patients.
Landscapes
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Virology (AREA)
- Health & Medical Sciences (AREA)
- Organic Chemistry (AREA)
- Biophysics (AREA)
- Biochemistry (AREA)
- General Health & Medical Sciences (AREA)
- Genetics & Genomics (AREA)
- Medicinal Chemistry (AREA)
- Molecular Biology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Immunology (AREA)
- Peptides Or Proteins (AREA)
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
EP22162453 | 2022-03-16 | ||
PCT/EP2023/056552 WO2023174976A1 (en) | 2022-03-16 | 2023-03-15 | Broadly neutralizing antibodies against hepatitis e virus |
Publications (1)
Publication Number | Publication Date |
---|---|
EP4493586A1 true EP4493586A1 (de) | 2025-01-22 |
Family
ID=80785039
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
EP23710899.8A Pending EP4493586A1 (de) | 2022-03-16 | 2023-03-15 | Breit neutralisierende antikörper gegen hepatitis-e-virus |
Country Status (2)
Country | Link |
---|---|
EP (1) | EP4493586A1 (de) |
WO (1) | WO2023174976A1 (de) |
Family Cites Families (6)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US5830636A (en) | 1993-12-22 | 1998-11-03 | Abbott Laboratories | HEV orf-2 monoclonal antibodies and methods for using same |
WO1995017501A1 (en) | 1993-12-22 | 1995-06-29 | Abbott Laboratories | Monoclonal antibodies against hev orf-2 and methods for using same |
BR122015016384B8 (pt) | 2001-11-08 | 2021-07-27 | Beijing Wantai Biological Pharmacy Entpr Co Ltd | polipeptídio recombinante ou isolado, molécula de ácido nucléico, kit de diagnóstico para detectar a infecção pelo vírus da hepatite e, composição de vacina, composição farmacêutica, vetor de expressão recombinante, célula hospedeira transformada e uso do polipeptídeo recombinante ou isolado |
CN110891966B (zh) | 2017-01-30 | 2024-02-06 | 国家医疗保健研究所 | 戊型肝炎病毒orf2衣壳多肽及其用途 |
WO2020011752A1 (en) * | 2018-07-10 | 2020-01-16 | INSERM (Institut National de la Santé et de la Recherche Médicale) | Antibodies having specificity for the orf2i protein of hepatitis e virus and uses thereof for diagnostic purposes |
ES2945571T3 (es) | 2018-07-10 | 2023-07-04 | Inst Nat Sante Rech Med | Anticuerpos que tienen especificidad por la proteína ORF2i del virus de la hepatitis E y usos de los mismos con fines de diagnóstico |
-
2023
- 2023-03-15 EP EP23710899.8A patent/EP4493586A1/de active Pending
- 2023-03-15 WO PCT/EP2023/056552 patent/WO2023174976A1/en active Application Filing
Also Published As
Publication number | Publication date |
---|---|
WO2023174976A1 (en) | 2023-09-21 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
CN112250763B (zh) | 靶向SARS-CoV-2冠状病毒的抗体及其诊断和检测用途 | |
WO2021196268A1 (zh) | 针对冠状病毒的具有中和活性的抗体及其用途 | |
US10906965B2 (en) | Methods of treating autoimmune disease or chronic inflammation wtih antibodies that bind peptidoglycan recognition protein 1 | |
US20240117011A1 (en) | Antibodies targeting the spike protein of coronaviruses | |
CA3164979A1 (en) | New polypeptide complex | |
CN113416245A (zh) | 一种可结合SARS-CoV-2病毒RBD蛋白的中和抗体及其应用 | |
US20240376178A1 (en) | Antibodies capable of binding to the spike protein of cononavirus sars-cov-2 | |
Ren et al. | The role of the light chain in the structure and binding activity of two cattle antibodies that neutralize bovine respiratory syncytial virus | |
BR112021005669A2 (pt) | anticorpos contra bcma solúvel | |
US11884718B2 (en) | Potent zika virus-specific and cross-neutralizing monoclonal antibodies to zika and dengue viruses following ZIKV infection or vaccination | |
WO2019156223A1 (ja) | チクングニアウイルスに対する抗体またはその抗原結合断片、およびその用途 | |
EP4493586A1 (de) | Breit neutralisierende antikörper gegen hepatitis-e-virus | |
US20230331820A1 (en) | HSV gE ANTIBODIES | |
CN111606992B (zh) | 一种抗呼吸道合胞病毒的全人源抗体 | |
EP3209697A1 (de) | Fn14-bindende proteine und verwendungen davon | |
US20230365658A1 (en) | REPAIRED THERAPEUTIC AND PROPHYLACTIC ANTIBODIES AGAINST SARS-CoV-2 VARIANTS | |
JP2025507595A (ja) | コロナウイルスSARS-CoV-2のスパイクタンパク質に結合することができる抗体 | |
WO2023240246A1 (en) | Computationally engineered monocolonal antibodies and antigen binding fragments specific for sars-cov-2 spike proteins and uses thereof | |
WO2024150074A2 (en) | Coronavirus antibodies and therapeutic uses thereof | |
TW202216758A (zh) | 抗人類免疫缺陷病毒-1抗體、包含其的細胞、核酸、組合物及套組 | |
WO2024058987A2 (en) | Polypeptides effective against multiple coronaviruses |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
STAA | Information on the status of an ep patent application or granted ep patent |
Free format text: STATUS: UNKNOWN |
|
STAA | Information on the status of an ep patent application or granted ep patent |
Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE |
|
PUAI | Public reference made under article 153(3) epc to a published international application that has entered the european phase |
Free format text: ORIGINAL CODE: 0009012 |
|
STAA | Information on the status of an ep patent application or granted ep patent |
Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE |
|
17P | Request for examination filed |
Effective date: 20240902 |
|
AK | Designated contracting states |
Kind code of ref document: A1 Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC ME MK MT NL NO PL PT RO RS SE SI SK SM TR |