WO2022079606A1 - Sars cov-2 neutralizing antibodies - Google Patents
Sars cov-2 neutralizing antibodies Download PDFInfo
- Publication number
- WO2022079606A1 WO2022079606A1 PCT/IB2021/059363 IB2021059363W WO2022079606A1 WO 2022079606 A1 WO2022079606 A1 WO 2022079606A1 IB 2021059363 W IB2021059363 W IB 2021059363W WO 2022079606 A1 WO2022079606 A1 WO 2022079606A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- seq
- cov
- antibodies
- sars
- antibody
- Prior art date
Links
- 230000003472 neutralizing effect Effects 0.000 title abstract description 22
- 230000027455 binding Effects 0.000 claims abstract description 51
- 241001678559 COVID-19 virus Species 0.000 claims abstract description 36
- 208000025721 COVID-19 Diseases 0.000 claims abstract description 23
- 108091005609 SARS-CoV-2 Spike Subunit S1 Proteins 0.000 claims abstract description 9
- 239000007922 nasal spray Substances 0.000 claims abstract description 6
- 229940097496 nasal spray Drugs 0.000 claims abstract description 6
- 239000000203 mixture Substances 0.000 claims description 25
- 208000024891 symptom Diseases 0.000 claims description 22
- 239000012634 fragment Substances 0.000 claims description 13
- 239000008194 pharmaceutical composition Substances 0.000 claims description 12
- 238000000034 method Methods 0.000 claims description 10
- 108020004707 nucleic acids Proteins 0.000 claims description 7
- 102000039446 nucleic acids Human genes 0.000 claims description 7
- 150000007523 nucleic acids Chemical class 0.000 claims description 7
- 230000010530 Virus Neutralization Effects 0.000 claims description 6
- 102000044437 S1 domains Human genes 0.000 claims description 5
- 239000003937 drug carrier Substances 0.000 claims description 5
- 108091005634 SARS-CoV-2 receptor-binding domains Proteins 0.000 claims description 4
- 241000282414 Homo sapiens Species 0.000 abstract description 31
- 230000009385 viral infection Effects 0.000 abstract description 8
- 208000036142 Viral infection Diseases 0.000 abstract description 7
- 210000004400 mucous membrane Anatomy 0.000 abstract description 4
- 108060003951 Immunoglobulin Proteins 0.000 abstract description 3
- 102000018358 immunoglobulin Human genes 0.000 abstract description 3
- 230000003449 preventive effect Effects 0.000 abstract description 2
- 229940126585 therapeutic drug Drugs 0.000 abstract description 2
- 210000004027 cell Anatomy 0.000 description 33
- 241000700605 Viruses Species 0.000 description 21
- 108090000623 proteins and genes Proteins 0.000 description 21
- 238000003556 assay Methods 0.000 description 18
- 102000004169 proteins and genes Human genes 0.000 description 16
- 201000010099 disease Diseases 0.000 description 14
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 14
- 238000011282 treatment Methods 0.000 description 14
- 101001028244 Onchocerca volvulus Fatty-acid and retinol-binding protein 1 Proteins 0.000 description 12
- 229960005486 vaccine Drugs 0.000 description 12
- 201000003176 Severe Acute Respiratory Syndrome Diseases 0.000 description 11
- 230000007423 decrease Effects 0.000 description 10
- 208000037847 SARS-CoV-2-infection Diseases 0.000 description 9
- 238000012575 bio-layer interferometry Methods 0.000 description 9
- 210000004602 germ cell Anatomy 0.000 description 9
- 208000015181 infectious disease Diseases 0.000 description 9
- 102100035360 Cerebellar degeneration-related antigen 1 Human genes 0.000 description 8
- 238000002965 ELISA Methods 0.000 description 8
- 238000006243 chemical reaction Methods 0.000 description 8
- 210000003819 peripheral blood mononuclear cell Anatomy 0.000 description 8
- 230000002441 reversible effect Effects 0.000 description 8
- 239000000427 antigen Substances 0.000 description 7
- 102000036639 antigens Human genes 0.000 description 7
- 108091007433 antigens Proteins 0.000 description 7
- 239000002299 complementary DNA Substances 0.000 description 7
- 241000711573 Coronaviridae Species 0.000 description 6
- 229940096437 Protein S Drugs 0.000 description 6
- 238000009472 formulation Methods 0.000 description 6
- 230000028993 immune response Effects 0.000 description 6
- 108090000765 processed proteins & peptides Proteins 0.000 description 6
- 230000001225 therapeutic effect Effects 0.000 description 6
- 101100454807 Caenorhabditis elegans lgg-1 gene Proteins 0.000 description 5
- 102100031673 Corneodesmosin Human genes 0.000 description 5
- 230000000903 blocking effect Effects 0.000 description 5
- 238000012512 characterization method Methods 0.000 description 5
- 230000000120 cytopathologic effect Effects 0.000 description 5
- 230000000694 effects Effects 0.000 description 5
- 239000002609 medium Substances 0.000 description 5
- 102000004196 processed proteins & peptides Human genes 0.000 description 5
- 230000000069 prophylactic effect Effects 0.000 description 5
- 102000005962 receptors Human genes 0.000 description 5
- 108020003175 receptors Proteins 0.000 description 5
- 230000028327 secretion Effects 0.000 description 5
- 230000003612 virological effect Effects 0.000 description 5
- 238000012300 Sequence Analysis Methods 0.000 description 4
- 108010031318 Vitronectin Proteins 0.000 description 4
- 238000010367 cloning Methods 0.000 description 4
- 239000003085 diluting agent Substances 0.000 description 4
- 244000052769 pathogen Species 0.000 description 4
- 239000013612 plasmid Substances 0.000 description 4
- 239000013598 vector Substances 0.000 description 4
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 3
- 102100035765 Angiotensin-converting enzyme 2 Human genes 0.000 description 3
- 108090000975 Angiotensin-converting enzyme 2 Proteins 0.000 description 3
- 101100454808 Caenorhabditis elegans lgg-2 gene Proteins 0.000 description 3
- 101100217502 Caenorhabditis elegans lgg-3 gene Proteins 0.000 description 3
- 208000035473 Communicable disease Diseases 0.000 description 3
- 108020004414 DNA Proteins 0.000 description 3
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N Dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 3
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 description 3
- 206010013975 Dyspnoeas Diseases 0.000 description 3
- 241000588724 Escherichia coli Species 0.000 description 3
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 3
- 108700036684 S1 domains Proteins 0.000 description 3
- 101000629318 Severe acute respiratory syndrome coronavirus 2 Spike glycoprotein Proteins 0.000 description 3
- 230000000890 antigenic effect Effects 0.000 description 3
- 239000000872 buffer Substances 0.000 description 3
- 210000004978 chinese hamster ovary cell Anatomy 0.000 description 3
- 150000001875 compounds Chemical class 0.000 description 3
- 238000001514 detection method Methods 0.000 description 3
- 230000036039 immunity Effects 0.000 description 3
- 238000011534 incubation Methods 0.000 description 3
- 239000007788 liquid Substances 0.000 description 3
- 239000000463 material Substances 0.000 description 3
- 238000006386 neutralization reaction Methods 0.000 description 3
- 230000001717 pathogenic effect Effects 0.000 description 3
- 239000000546 pharmaceutical excipient Substances 0.000 description 3
- 239000002953 phosphate buffered saline Substances 0.000 description 3
- 238000002415 sodium dodecyl sulfate polyacrylamide gel electrophoresis Methods 0.000 description 3
- 239000000243 solution Substances 0.000 description 3
- 102100030988 Angiotensin-converting enzyme Human genes 0.000 description 2
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 2
- 108091026890 Coding region Proteins 0.000 description 2
- 208000000059 Dyspnea Diseases 0.000 description 2
- 102000004190 Enzymes Human genes 0.000 description 2
- 108090000790 Enzymes Proteins 0.000 description 2
- 108090000288 Glycoproteins Proteins 0.000 description 2
- 102000003886 Glycoproteins Human genes 0.000 description 2
- 241001465754 Metazoa Species 0.000 description 2
- 108010076504 Protein Sorting Signals Proteins 0.000 description 2
- 239000012980 RPMI-1640 medium Substances 0.000 description 2
- 241000315672 SARS coronavirus Species 0.000 description 2
- PXIPVTKHYLBLMZ-UHFFFAOYSA-N Sodium azide Chemical compound [Na+].[N-]=[N+]=[N-] PXIPVTKHYLBLMZ-UHFFFAOYSA-N 0.000 description 2
- 101710198474 Spike protein Proteins 0.000 description 2
- 108010067390 Viral Proteins Proteins 0.000 description 2
- 230000002411 adverse Effects 0.000 description 2
- 238000012436 analytical size exclusion chromatography Methods 0.000 description 2
- 238000013459 approach Methods 0.000 description 2
- 210000003719 b-lymphocyte Anatomy 0.000 description 2
- 230000001580 bacterial effect Effects 0.000 description 2
- 230000009286 beneficial effect Effects 0.000 description 2
- 230000004071 biological effect Effects 0.000 description 2
- 238000010804 cDNA synthesis Methods 0.000 description 2
- 210000000170 cell membrane Anatomy 0.000 description 2
- 230000003833 cell viability Effects 0.000 description 2
- 238000011161 development Methods 0.000 description 2
- 238000010790 dilution Methods 0.000 description 2
- 239000012895 dilution Substances 0.000 description 2
- 239000003814 drug Substances 0.000 description 2
- 238000005516 engineering process Methods 0.000 description 2
- 238000002474 experimental method Methods 0.000 description 2
- 239000013604 expression vector Substances 0.000 description 2
- 239000012091 fetal bovine serum Substances 0.000 description 2
- 239000000499 gel Substances 0.000 description 2
- 230000005764 inhibitory process Effects 0.000 description 2
- 238000002955 isolation Methods 0.000 description 2
- 230000000670 limiting effect Effects 0.000 description 2
- 210000004072 lung Anatomy 0.000 description 2
- 238000004519 manufacturing process Methods 0.000 description 2
- 230000035772 mutation Effects 0.000 description 2
- 230000036961 partial effect Effects 0.000 description 2
- 229920001184 polypeptide Polymers 0.000 description 2
- 230000003389 potentiating effect Effects 0.000 description 2
- 238000002360 preparation method Methods 0.000 description 2
- 230000002265 prevention Effects 0.000 description 2
- 238000012545 processing Methods 0.000 description 2
- 239000000047 product Substances 0.000 description 2
- 230000000644 propagated effect Effects 0.000 description 2
- 230000001681 protective effect Effects 0.000 description 2
- 230000009467 reduction Effects 0.000 description 2
- 230000002829 reductive effect Effects 0.000 description 2
- 210000002345 respiratory system Anatomy 0.000 description 2
- 208000013220 shortness of breath Diseases 0.000 description 2
- 239000007921 spray Substances 0.000 description 2
- UCSJYZPVAKXKNQ-HZYVHMACSA-N streptomycin Chemical compound CN[C@H]1[C@H](O)[C@@H](O)[C@H](CO)O[C@H]1O[C@@H]1[C@](C=O)(O)[C@H](C)O[C@H]1O[C@@H]1[C@@H](NC(N)=N)[C@H](O)[C@@H](NC(N)=N)[C@H](O)[C@H]1O UCSJYZPVAKXKNQ-HZYVHMACSA-N 0.000 description 2
- 210000001519 tissue Anatomy 0.000 description 2
- 239000003981 vehicle Substances 0.000 description 2
- 244000052613 viral pathogen Species 0.000 description 2
- 102000040650 (ribonucleotides)n+m Human genes 0.000 description 1
- FWMNVWWHGCHHJJ-SKKKGAJSSA-N 4-amino-1-[(2r)-6-amino-2-[[(2r)-2-[[(2r)-2-[[(2r)-2-amino-3-phenylpropanoyl]amino]-3-phenylpropanoyl]amino]-4-methylpentanoyl]amino]hexanoyl]piperidine-4-carboxylic acid Chemical compound C([C@H](C(=O)N[C@H](CC(C)C)C(=O)N[C@H](CCCCN)C(=O)N1CCC(N)(CC1)C(O)=O)NC(=O)[C@H](N)CC=1C=CC=CC=1)C1=CC=CC=C1 FWMNVWWHGCHHJJ-SKKKGAJSSA-N 0.000 description 1
- 102400000344 Angiotensin-1 Human genes 0.000 description 1
- 101800000734 Angiotensin-1 Proteins 0.000 description 1
- 101100272788 Arabidopsis thaliana BSL3 gene Proteins 0.000 description 1
- 241000894006 Bacteria Species 0.000 description 1
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 1
- OYPRJOBELJOOCE-UHFFFAOYSA-N Calcium Chemical compound [Ca] OYPRJOBELJOOCE-UHFFFAOYSA-N 0.000 description 1
- 206010008479 Chest Pain Diseases 0.000 description 1
- 241000288673 Chiroptera Species 0.000 description 1
- 108020004705 Codon Proteins 0.000 description 1
- 108020004635 Complementary DNA Proteins 0.000 description 1
- 101710139375 Corneodesmosin Proteins 0.000 description 1
- 206010011224 Cough Diseases 0.000 description 1
- 206010061818 Disease progression Diseases 0.000 description 1
- 238000012286 ELISA Assay Methods 0.000 description 1
- WQZGKKKJIJFFOK-GASJEMHNSA-N Glucose Chemical compound OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 1
- 239000012981 Hank's balanced salt solution Substances 0.000 description 1
- 206010019233 Headaches Diseases 0.000 description 1
- 241000282412 Homo Species 0.000 description 1
- 101000839660 Homo sapiens Immunoglobulin heavy variable 3-53 Proteins 0.000 description 1
- 101000839658 Homo sapiens Immunoglobulin heavy variable 3-66 Proteins 0.000 description 1
- 101001047626 Homo sapiens Immunoglobulin kappa variable 2-24 Proteins 0.000 description 1
- 101000956885 Homo sapiens Immunoglobulin lambda variable 2-14 Proteins 0.000 description 1
- 206010061598 Immunodeficiency Diseases 0.000 description 1
- 102000006496 Immunoglobulin Heavy Chains Human genes 0.000 description 1
- 108010019476 Immunoglobulin Heavy Chains Proteins 0.000 description 1
- 102000013463 Immunoglobulin Light Chains Human genes 0.000 description 1
- 108010065825 Immunoglobulin Light Chains Proteins 0.000 description 1
- 102100028317 Immunoglobulin heavy variable 3-53 Human genes 0.000 description 1
- 102100027821 Immunoglobulin heavy variable 3-66 Human genes 0.000 description 1
- 102100022947 Immunoglobulin kappa variable 2-24 Human genes 0.000 description 1
- 102100038429 Immunoglobulin lambda variable 2-14 Human genes 0.000 description 1
- 241000712431 Influenza A virus Species 0.000 description 1
- 108091092195 Intron Proteins 0.000 description 1
- 102000003960 Ligases Human genes 0.000 description 1
- 108090000364 Ligases Proteins 0.000 description 1
- FYYHWMGAXLPEAU-UHFFFAOYSA-N Magnesium Chemical compound [Mg] FYYHWMGAXLPEAU-UHFFFAOYSA-N 0.000 description 1
- 208000000112 Myalgia Diseases 0.000 description 1
- 108091028043 Nucleic acid sequence Proteins 0.000 description 1
- 206010068319 Oropharyngeal pain Diseases 0.000 description 1
- 229930182555 Penicillin Natural products 0.000 description 1
- JGSARLDLIJGVTE-MBNYWOFBSA-N Penicillin G Chemical compound N([C@H]1[C@H]2SC([C@@H](N2C1=O)C(O)=O)(C)C)C(=O)CC1=CC=CC=C1 JGSARLDLIJGVTE-MBNYWOFBSA-N 0.000 description 1
- 201000007100 Pharyngitis Diseases 0.000 description 1
- 206010035664 Pneumonia Diseases 0.000 description 1
- 206010037660 Pyrexia Diseases 0.000 description 1
- 238000002123 RNA extraction Methods 0.000 description 1
- 208000036071 Rhinorrhea Diseases 0.000 description 1
- 206010039101 Rhinorrhoea Diseases 0.000 description 1
- 241001678561 Sarbecovirus Species 0.000 description 1
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 1
- 101150003725 TK gene Proteins 0.000 description 1
- 238000012338 Therapeutic targeting Methods 0.000 description 1
- 239000007983 Tris buffer Substances 0.000 description 1
- 206010053614 Type III immune complex mediated reaction Diseases 0.000 description 1
- 239000004480 active ingredient Substances 0.000 description 1
- 239000013543 active substance Substances 0.000 description 1
- 230000001154 acute effect Effects 0.000 description 1
- 239000000654 additive Substances 0.000 description 1
- 230000000996 additive effect Effects 0.000 description 1
- 238000001261 affinity purification Methods 0.000 description 1
- 208000026935 allergic disease Diseases 0.000 description 1
- AVKUERGKIZMTKX-NJBDSQKTSA-N ampicillin Chemical compound C1([C@@H](N)C(=O)N[C@H]2[C@H]3SC([C@@H](N3C2=O)C(O)=O)(C)C)=CC=CC=C1 AVKUERGKIZMTKX-NJBDSQKTSA-N 0.000 description 1
- 229960000723 ampicillin Drugs 0.000 description 1
- 230000003321 amplification Effects 0.000 description 1
- 238000004458 analytical method Methods 0.000 description 1
- ORWYRWWVDCYOMK-HBZPZAIKSA-N angiotensin I Chemical compound C([C@@H](C(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@@H](CC=1NC=NC=1)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](CC=1NC=NC=1)C(=O)N[C@@H](CC(C)C)C(O)=O)NC(=O)[C@@H](NC(=O)[C@H](CCCN=C(N)N)NC(=O)[C@@H](N)CC(O)=O)C(C)C)C1=CC=C(O)C=C1 ORWYRWWVDCYOMK-HBZPZAIKSA-N 0.000 description 1
- 239000003443 antiviral agent Substances 0.000 description 1
- 210000001367 artery Anatomy 0.000 description 1
- 239000012298 atmosphere Substances 0.000 description 1
- 230000008901 benefit Effects 0.000 description 1
- 238000013357 binding ELISA Methods 0.000 description 1
- 230000005540 biological transmission Effects 0.000 description 1
- 230000015572 biosynthetic process Effects 0.000 description 1
- 210000004369 blood Anatomy 0.000 description 1
- 239000008280 blood Substances 0.000 description 1
- UDSAIICHUKSCKT-UHFFFAOYSA-N bromophenol blue Chemical compound C1=C(Br)C(O)=C(Br)C=C1C1(C=2C=C(Br)C(O)=C(Br)C=2)C2=CC=CC=C2S(=O)(=O)O1 UDSAIICHUKSCKT-UHFFFAOYSA-N 0.000 description 1
- 229910052791 calcium Inorganic materials 0.000 description 1
- 239000011575 calcium Substances 0.000 description 1
- 238000004113 cell culture Methods 0.000 description 1
- 239000006285 cell suspension Substances 0.000 description 1
- 238000012054 celltiter-glo Methods 0.000 description 1
- 239000003638 chemical reducing agent Substances 0.000 description 1
- 239000003795 chemical substances by application Substances 0.000 description 1
- 239000013599 cloning vector Substances 0.000 description 1
- 230000000052 comparative effect Effects 0.000 description 1
- 230000024203 complement activation Effects 0.000 description 1
- 238000010276 construction Methods 0.000 description 1
- 239000012228 culture supernatant Substances 0.000 description 1
- 230000003247 decreasing effect Effects 0.000 description 1
- 230000007123 defense Effects 0.000 description 1
- 230000003111 delayed effect Effects 0.000 description 1
- 230000001419 dependent effect Effects 0.000 description 1
- 238000013461 design Methods 0.000 description 1
- 238000003745 diagnosis Methods 0.000 description 1
- LOKCTEFSRHRXRJ-UHFFFAOYSA-I dipotassium trisodium dihydrogen phosphate hydrogen phosphate dichloride Chemical compound P(=O)(O)(O)[O-].[K+].P(=O)(O)([O-])[O-].[Na+].[Na+].[Cl-].[K+].[Cl-].[Na+] LOKCTEFSRHRXRJ-UHFFFAOYSA-I 0.000 description 1
- 238000007598 dipping method Methods 0.000 description 1
- 230000005750 disease progression Effects 0.000 description 1
- 239000012153 distilled water Substances 0.000 description 1
- 230000009429 distress Effects 0.000 description 1
- 239000002552 dosage form Substances 0.000 description 1
- 229940079593 drug Drugs 0.000 description 1
- 230000004064 dysfunction Effects 0.000 description 1
- 239000012636 effector Substances 0.000 description 1
- 230000012202 endocytosis Effects 0.000 description 1
- 210000001163 endosome Anatomy 0.000 description 1
- 230000026502 entry into host cell Effects 0.000 description 1
- 230000002255 enzymatic effect Effects 0.000 description 1
- 239000000945 filler Substances 0.000 description 1
- 239000012467 final product Substances 0.000 description 1
- 238000007667 floating Methods 0.000 description 1
- 239000006260 foam Substances 0.000 description 1
- 238000012395 formulation development Methods 0.000 description 1
- 230000006870 function Effects 0.000 description 1
- 230000005182 global health Effects 0.000 description 1
- 239000001963 growth medium Substances 0.000 description 1
- 231100000869 headache Toxicity 0.000 description 1
- 210000002216 heart Anatomy 0.000 description 1
- 238000010438 heat treatment Methods 0.000 description 1
- 230000016178 immune complex formation Effects 0.000 description 1
- 238000002649 immunization Methods 0.000 description 1
- 230000003053 immunization Effects 0.000 description 1
- 230000006872 improvement Effects 0.000 description 1
- 206010022000 influenza Diseases 0.000 description 1
- 239000004615 ingredient Substances 0.000 description 1
- 230000000266 injurious effect Effects 0.000 description 1
- 238000003780 insertion Methods 0.000 description 1
- 230000037431 insertion Effects 0.000 description 1
- 230000003993 interaction Effects 0.000 description 1
- 238000005305 interferometry Methods 0.000 description 1
- 210000000936 intestine Anatomy 0.000 description 1
- 238000001990 intravenous administration Methods 0.000 description 1
- 230000007794 irritation Effects 0.000 description 1
- 238000005304 joining Methods 0.000 description 1
- 210000003734 kidney Anatomy 0.000 description 1
- 238000012933 kinetic analysis Methods 0.000 description 1
- 239000012160 loading buffer Substances 0.000 description 1
- 230000007774 longterm Effects 0.000 description 1
- 239000000314 lubricant Substances 0.000 description 1
- 210000002540 macrophage Anatomy 0.000 description 1
- 229910052749 magnesium Inorganic materials 0.000 description 1
- 239000011777 magnesium Substances 0.000 description 1
- 239000003550 marker Substances 0.000 description 1
- 230000034217 membrane fusion Effects 0.000 description 1
- 238000002625 monoclonal antibody therapy Methods 0.000 description 1
- 210000003928 nasal cavity Anatomy 0.000 description 1
- 229940052404 nasal powder Drugs 0.000 description 1
- 210000001989 nasopharynx Anatomy 0.000 description 1
- 229910052757 nitrogen Inorganic materials 0.000 description 1
- 238000003199 nucleic acid amplification method Methods 0.000 description 1
- 230000000474 nursing effect Effects 0.000 description 1
- QIQXTHQIDYTFRH-UHFFFAOYSA-N octadecanoic acid Chemical compound CCCCCCCCCCCCCCCCCC(O)=O QIQXTHQIDYTFRH-UHFFFAOYSA-N 0.000 description 1
- 239000002674 ointment Substances 0.000 description 1
- 210000000056 organ Anatomy 0.000 description 1
- 238000012856 packing Methods 0.000 description 1
- 238000002638 palliative care Methods 0.000 description 1
- 238000004091 panning Methods 0.000 description 1
- 239000003182 parenteral nutrition solution Substances 0.000 description 1
- 239000002245 particle Substances 0.000 description 1
- 229940049954 penicillin Drugs 0.000 description 1
- 230000003169 placental effect Effects 0.000 description 1
- 239000004033 plastic Substances 0.000 description 1
- 210000004043 pneumocyte Anatomy 0.000 description 1
- 108091033319 polynucleotide Proteins 0.000 description 1
- 102000040430 polynucleotide Human genes 0.000 description 1
- 239000002157 polynucleotide Substances 0.000 description 1
- 230000008569 process Effects 0.000 description 1
- 230000002250 progressing effect Effects 0.000 description 1
- 230000001737 promoting effect Effects 0.000 description 1
- 238000011321 prophylaxis Methods 0.000 description 1
- 238000000159 protein binding assay Methods 0.000 description 1
- 238000000746 purification Methods 0.000 description 1
- 238000011160 research Methods 0.000 description 1
- 230000000241 respiratory effect Effects 0.000 description 1
- 230000004044 response Effects 0.000 description 1
- 238000003757 reverse transcription PCR Methods 0.000 description 1
- 238000012216 screening Methods 0.000 description 1
- 238000000926 separation method Methods 0.000 description 1
- 238000012163 sequencing technique Methods 0.000 description 1
- 239000011780 sodium chloride Substances 0.000 description 1
- 239000007787 solid Substances 0.000 description 1
- 239000002904 solvent Substances 0.000 description 1
- 238000003860 storage Methods 0.000 description 1
- 229960005322 streptomycin Drugs 0.000 description 1
- 239000006228 supernatant Substances 0.000 description 1
- 239000000725 suspension Substances 0.000 description 1
- 230000002195 synergetic effect Effects 0.000 description 1
- 238000003786 synthesis reaction Methods 0.000 description 1
- 230000009885 systemic effect Effects 0.000 description 1
- 239000000454 talc Substances 0.000 description 1
- 229910052623 talc Inorganic materials 0.000 description 1
- 230000008685 targeting Effects 0.000 description 1
- 229940124597 therapeutic agent Drugs 0.000 description 1
- 238000002560 therapeutic procedure Methods 0.000 description 1
- 230000003867 tiredness Effects 0.000 description 1
- 208000016255 tiredness Diseases 0.000 description 1
- 230000001988 toxicity Effects 0.000 description 1
- 231100000419 toxicity Toxicity 0.000 description 1
- 238000001890 transfection Methods 0.000 description 1
- 230000009466 transformation Effects 0.000 description 1
- 230000005945 translocation Effects 0.000 description 1
- 108091005703 transmembrane proteins Proteins 0.000 description 1
- 102000035160 transmembrane proteins Human genes 0.000 description 1
- 230000032258 transport Effects 0.000 description 1
- LENZDBCJOHFCAS-UHFFFAOYSA-N tris Chemical compound OCC(N)(CO)CO LENZDBCJOHFCAS-UHFFFAOYSA-N 0.000 description 1
- 241001529453 unidentified herpesvirus Species 0.000 description 1
- 238000002255 vaccination Methods 0.000 description 1
- 229940125575 vaccine candidate Drugs 0.000 description 1
- 230000035899 viability Effects 0.000 description 1
- 238000005406 washing Methods 0.000 description 1
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Chemical compound O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 1
- 238000001262 western blot Methods 0.000 description 1
- XOOUIPVCVHRTMJ-UHFFFAOYSA-L zinc stearate Chemical compound [Zn+2].CCCCCCCCCCCCCCCCCC([O-])=O.CCCCCCCCCCCCCCCCCC([O-])=O XOOUIPVCVHRTMJ-UHFFFAOYSA-L 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/12—Antivirals
- A61P31/14—Antivirals for RNA viruses
-
- 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
- C07K16/1002—Coronaviridae
- C07K16/1003—Severe acute respiratory syndrome coronavirus 2 [SARS‐CoV‐2 or Covid-19]
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/20—Immunoglobulins specific features characterized by taxonomic origin
- C07K2317/21—Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
-
- 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
- This application contains a sequence listing provided in computer readable format.
- This application relates to novel antibodies and specifically to novel SARS CoV-2 neutralizing antibodies.
- COVID 19-pandemic is a pandemic of coronavirus disease 2019 caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2).
- SARS-CoV-2 is a novel virus infecting human beings: first human cases of COVID-19 were identified in Wuhan, China, in December 2019.
- the virus has infected more than 33 million people and at least 998,200 people have died.
- the virus has been detected in nearly every country.
- the novel coronavirus has had devastating global health, as well as economic consequences and even if there are several vaccine candidates under development the treatment and prevention of the viral infection are still to be developed.
- ACE2 angiotensin I and converting enzyme 2
- SARS-CoV-2 angiotensin I and converting enzyme 2
- ACE2 is an enzyme attached to the cell membranes of cells located in the lungs, arteries, heart, kidney, and intestines.
- ACE2 serves as the main entry point into cells for some coronaviruses, including SARS-CoV-2. More specifically, the binding of the spike S1 protein of SARS-CoV-2 to the enzymatic domain of ACE2 on the surface of cells results in endocytosis and translocation of both the virus and the enzyme into endosomes located within cells.
- Neutralizing antibodies can block the entry of a pathogen into the cell and thus prevent the pathogen from infecting the body.
- ACE2 is the main entry point for SARS-Cov-2, finding efficient neutralizing antibodies that could block the entry seems a promising approach for developing prophylactic and therapeutic means to fight against the pandemic.
- the present invention provides neutralizing antibodies and antibody compositions that are suitable for administration to a subject having SARS-Cov-2 infection, or at risk of SARS-Cov-2 infection, in an amount and according to a schedule sufficient to prevent or reduce the virus infection.
- the present invention provides antibodies suitable for use as a passive vaccine against the SARS-CoV-2 comprising at least one antibody of the invention and a pharmaceutically acceptable carrier. According to certain aspects, the present invention provides antibodies suitable for use as a nasal spray to provide prophylactic treatment against SARS-CoV-2 infection.
- the present invention provides an antibody-based pharmaceutical composition comprising an effective amount of one of more of the isolated SARS-CoV-2 antibodies described herein for providing a prophylactic or therapeutic treatment to reduce infection of the SARS-CoV- 2.
- VH heavy chain variable region
- VL light chain variable region
- the isolated SARS-CoV-2 S1 -domain binding antibody or fragment thereof wherein the antibody comprises the VH protein according to SEQ ID NO: 44 or 72 and VL protein according to SEQ ID NO: 48 or 74, respectively.
- It is an object of this invention to provide a composition comprising the isolated SARS-CoV-2 S1 -domain binding antibody comprising a heavy chain variable region (VH) comprising the VH CDR 1 -3 according to SEQ ID Nos: 95-97, respectively, and a light chain variable region (VL) comprising the VL CDR1 -3 of SEQ ID Nos: 89-91 , respectively.
- VH heavy chain variable region
- VL light chain variable region
- composition additionally comprises one or more of the antibodies comprising a heavy chain variable region VH according to SEQ ID NO: 60 and light chain variable region VL according to SEQ ID NO: 62, or VH according to SEQ ID NO: 64 and VL according to SEQ ID NO: 66, or VH according to SEQ ID NO: 68 and VL according to SEQ ID NO: 70.
- composition is a pharmaceutical composition, comprising a pharmaceutically acceptable carrier.
- composition is a nasal spray.
- FIG 1 illustrates the infection cycle of SARS-CoV-2.
- SARS-CoV-2 uses angiotensin converting enzyme 2 (ACE2) receptor to enter the host cell in which the virus then replicates.
- ACE2 angiotensin converting enzyme 2
- Virus inserts its RNA into the host cell, where it utilizes the host cell to produce new viral proteins and replicate the RNA genome.
- Viral proteins are glycosylated and transported to the host cell membrane, where the assembled particles bud from the host cell.
- Neutralizing antibodies can bind to spike S1 protein and thereby prevent or impair its capability to bind to ACE-2 receptor.
- Figure 2a-d shows analytical Size-Exclusion Chromatography of SARS-CoV-2 neutralizing antibody clones, a) clone 23G7; b) clone 23E2, c) clone 8A12, d) clone 42B7.
- Figure 3A-D A) shows binding sensograms of SARS CoV-2 antibodies: a) clone 8A12 sample 12#22; b) clone 23E2, sample #81 ; c) clone 23G7, sample #86, d) clone 42B7, sample #140 to the S1 -domain of SARS-Cov-2;
- Figure 4 shows in-tandem bio-layer interferometry binning assay of SARS-CoV-2 RBD binding antibodies indicating the RBD binding antibodies belong to three independent epitope bins.
- Figure 5a-d shows ACE2 blocking assay by bio-layer interferometry, a) shows ACE2 binding of clone 8A12 antibodies; b) shows ACE2 binding of clone 23G7 antibodies; c) shows ACE 2 binding of clone 23E2 antibodies; d) shows ACE2 binding of 42B7 antibodies. All tested antibodies except 8A12 is able to block ACE2 binding.
- Figure 6 shows ACE binging inhibition assay by ELISA. Samples from Clones 23G7, 23E2, 8A12 and 42B7 were tested. Various concentrations of antibodies were used to treat immobilized S-protein followed by detection with biotinylated ACE2 and Strepavidin-HRP.
- Figure 7A-B A) shows SARS CoV-2 live virus neutralization assay. VERO E6 cells were treated with hCoV-19/Norway/Trondheim-E9/2020 at moi 0.1 together with various concentrations of SARS-CoV-2 antibodies of clones 8A12, 23G7 and 23E2, respectively. Cell viability was measured 72hours post-infection. B) shows live virus assay results measuring final antibody concentration where no cytopathic effects could be detected. Detailed description of the invention
- antibody is a polypeptide that specifically binds and recognizes an antigen such as S protein of SARS-Cov2 virus or an antigenic fragment thereof.
- a “monoclonal antibody” is an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigenic epitope.
- Vaccine as used here means a pharmaceutical composition that induces a prophylactic or therapeutic immune response in a subject.
- the immune response is a protective immune response.
- a vaccine induces an antigen-specific immune response to an antigen of a pathogen, for example a viral pathogen, such as SARS-Cov-2.
- a vaccine may include a polynucleotide (such as a nucleic acid encoding a disclosed antigen), or a peptide or polypeptide (such as a disclosed antigen).
- a vaccine induces an immune response that reduces the severity of the symptoms associated with SARS-Cov 2 infection and/or decreases the viral load compared to a control.
- a vaccine induces an immune response that reduces and/or prevents SARS-CoV-2 infection compared to a control.
- disease refers to any alternation in state of the body or of some of the organs, interrupting or disturbing the performance of the functions and/or causing symptoms such as discomfort, dysfunction, distress, or even death to the person afflicted or those in contact with a person.
- the term "in need thereof” means having a disease, being diagnosed with a disease, or being in need of preventing a disease.
- a subject in need thereof can be a subject in need of treating or preventing a disease.
- the terms “treat,” “treatment,” “treating,” refer to therapeutic treatments, wherein the object is to reverse, alleviate, ameliorate, inhibit, slow down or stop the progression or severity of an infectious disease such as COVID 19- disease.
- the term “treating” includes reducing or alleviating at least one adverse effect or symptom of an infectious disease. Treatment is generally “effective” if one or more symptoms or clinical markers are reduced. Alternatively, treatment is “effective” if the progression of a disease is reduced or halted.
- treatment includes not just the improvement of symptoms or markers, but also a cessation of, or at least slowing of, progress or worsening of symptoms compared to what would be expected in the absence of treatment.
- Beneficial or desired clinical results include, but are not limited to, alleviation of one or more symptoms, diminishment of extent of disease, stabilized (i.e. , not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, remission (whether partial or total), and/or decreased mortality.
- treatment is considered effective if the condition is stabilized, or the progression of COVID 19 disease is slowed or halted.
- treatment also includes providing relief from the symptoms or side-effects of the disease (including palliative treatment).
- prevent refers to stopping, hindering, and/or slowing down an infectious disease.
- prevention is synonymous with “inhibit”.
- administering refers to the placement of a compound as disclosed herein into a subject by a method or route which results in at least partial delivery of the agent at a desired site.
- Pharmaceutical compositions comprising the compounds disclosed herein can be administered by any appropriate route which results in an effective treatment in the subject.
- a "subject” means a human.
- the terms, “individual,” “patient” and “subject” are used interchangeably herein.
- the terms “increased”, “increase”, or “enhance” are all used herein to generally mean an increase by a statically significant amount; the terms “increased”, “increase”, or “enhance”, mean an increase of at least 10% as compared to a reference level, for example an increase of at least about 10%, at least about 20%, or at least about 30%, or at least about 40%, or at least about 50%, or at least about 60%, or at least about 70%, or at least about 80%, or at least about 90% or up to and including a 100% increase or any increase between 10-100% as compared to a reference level, or at least about a 2-fold, or at least about a 3-fold, or at least about a 4-fold, or at least about a 5-fold or at least about a 10-fold increase, or any increase between 2-fold and 10-fold or greater as compared to a reference level.
- “decrease”, “reduce”, “reduction”, or “inhibit” are all used herein generally to mean a decrease by a statistically significant amount.
- “decrease”, “reduce”, “reduction”, or “inhibit” means a decrease by at least 10% as compared to a reference level, for example a decrease by at least about 20%, or at least about 30%, or at least about 40%, or at least about 50%, or at least about 60%, or at least about 70%, or at least about 80%, or at least about 90% or up to and including a 100% decrease (e.g. absent level or non-detectable level as compared to a reference level), or any decrease between 10-100% as compared to a reference level.
- a marker or symptom in the context of a marker or symptom is meant a statistically significant decrease in such level.
- the decrease can be, for example, at least 10%, at least 20%, at least 30%, at least 40% or more, and is preferably down to a level accepted as within the range of normal for an individual without a given disease.
- the term "pharmaceutically acceptable” refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
- the term "pharmaceutically acceptable carrier” means a pharmaceutically-acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, manufacturing aid (e.g., lubricant, talc magnesium, calcium or zinc stearate, or steric acid), or solvent encapsulating material necessary or used in formulating an active ingredient or agent for delivery to a subject.
- a pharmaceutically-acceptable material such as a liquid or solid filler, diluent, excipient, manufacturing aid (e.g., lubricant, talc magnesium, calcium or zinc stearate, or steric acid), or solvent encapsulating material necessary or used in formulating an active ingredient or agent for delivery to a subject.
- manufacturing aid e.g., lubricant, talc magnesium, calcium or zinc stearate, or steric acid
- solvent encapsulating material necessary or used in formulating an active ingredient or agent for delivery to a subject.
- the term "in combination” refers to the use of more than one prophylactic and/or therapeutic agent simultaneously or sequentially and in a manner that their respective effects are additive or synergistic.
- composition as used herein may be a single or a combination of antibodies disclosed herein, which can be the same or different, in order to prophylactically or therapeutically treat the progression SARS Cov-2 infection after administration. Such combinations can be selected according to the desired immunity or effect.
- the antibody-based pharmaceutical composition of the present invention may be formulated by any number of strategies known in the art (e.g., see McGoff and Scher, 2000, Solution Formulation of Proteins/Peptides: In McNally, E. J., ed. Protein Formulation and Delivery. New York, N.Y.: Marcel Dekker; pp. 139-158; Akers and Defilippis, 2000, Peptides and Proteins as Parenteral Solutions. In: Pharmaceutical Formulation Development of Peptides and Proteins. Philadelphia, Pa.: Talyor and Francis; pp. 145-177; Akers, et al., 2002, Pharm. Biotechnol. 14:47-127).
- SARS-CoV-2 Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) emerged at the end of 2019 and was shortly thereafter sequenced.
- SARS-CoV-2 appears to have originated in bats from which closely related viruses and viral sequences have been identified.
- SARS-CoV-2 belongs to the sarbecovirus subgenus and is closely related to SARS-CoV which was responsible for an epidemic in 2002-2003 resulting in 8,098 cases and 774 fatalities worldwide.
- the lack of pre-existing immunity to SARS-CoV- 2 due to its divergence from the circulating endemic coronaviruses, and its efficient human-to-human transmission has resulted in the ongoing COVID-19 pandemic which has already caused more than 33 million infections and over million fatalities as of late-September 2020.
- SARS-CoV-2 infection starts upon attachment of the viral trimeric transmembrane spike (S) glycoprotein via a receptor-binding domain (RBD) to angiotensin-converting enzyme 2 glycoprotein (ACE2), leading to membrane fusion and entry into host cells c.f. Figure 1 .
- SARS-CoV- 2 spike protein is the main target of neutralizing antibodies (Abs) and a focus of vaccine design and therapeutic targeting strategies.
- Prophylactic and/or therapeutic anti-viral drugs could allow overcome the gap before safe and efficient vaccines become widely available and will continue to be useful in unvaccinated individuals or those who respond poorly to vaccination.
- a treatment that takes whole plasma from a recovered person, and infuses it into another patient with the disease monoclonal antibody therapy uses a much more targeted approach, specifically neutralizing SARS-CoV-2 infectivity. Therefore, SARS-CoV-2- targeting antibodies are really valuable as they are supposed to be an important bridge to a vaccine and a hedge in the event vaccines are delayed or work inefficiently.
- COVID-19 symptoms can range from very mild to severe. Some people may have only a few symptoms, and some people may have no symptoms at all. Some people may experience worsened symptoms, such as worsened shortness of breath and pneumonia, about a week after symptoms start.
- This invention provides novel neutralizing anti-SARS-CoV-2 antibodies.
- the invention is now described with examples and references to the figures.
- PBMC peripheral blood mononuclear cell
- Venous blood samples were collected and PBMCs were separated into BD Vacutainer® CPTTM Cell Preparation Tubes containing the cell separation medium (BD Biosciences, Cat No.362753) according to manufacturer’s instructions.
- the collected PBMCs were stored under liquid nitrogen up to further processing for the isolation of the monoclonal antibodies.
- Monoclonal antibodies recognizing SARS-Cov-2 S1 protein were isolated using Hybrifree technology (Kivi G, et al., BMC Biotechnol. 2016; 16:2 incorporated herein by reference). Briefly, the frozen PBMCs was thawed, washed and collected into 10 ml of RPMI1640 supplemented with penicillin/streptomycin and 10 % of heat inactivated fetal bovine serum. Then the cells were seeded into a 100 mm cell culture dish and incubated ⁇ 1 h at 37 °C in an 8 % CO2 atmosphere. Then, free- floating cells (the fraction enriched for B-cells and separated from plastic-adherent cells, e.g. macrophages) were collected, viable cell count was determined and the cells were transferred into the capture medium (RPMI1640 supplemented with 0.5 % BSA and 0.1 % NaN 3 ).
- RNA isolation and cDNA synthesis were lysed and subjected to total RNA isolation and cDNA synthesis using oligo-T primer.
- the cDNA synthesis was conducted using SuperScriptTM IV First-Strand Synthesis System (Thermo Fischer Scientific Catalog number: 18091050) and oligo d(T)2o primer supplied with the kit.
- the cDNAs of antibody VH and VL regions were amplified using cocktails of forward primers that bind to FR1 regions of VH or VL kappa and VLJambda light chain coding sequences and were designed to maximally cover the variability of the human VH and VL sequences.
- single reverse primer was used that binds to the beginning of the constant region encoding sequence of the human IgG heavy chains, kappa light chain or lambda light chain, respectively.
- VH forward primers (5’-3’) were according to SEQ ID NO:9 - SEQ ID NO: 22.
- the VH reverse primer (5’-3’) was according to SEQ ID NO:23.
- VL kappa forward primers (5’- 3’) were according to SEQ ID NO 24-33.
- VL kappa reverse primer was according to SEQ ID NO:34.
- VL lambda forward primers (5’-3’) were according to SEQ ID NO: 35-41 and SEQ ID NO:98.
- the VL lambda reverse primer was according to SEQ ID NO: 42.
- VH and VL PCR products retrieved from the same capture reaction were both exactly joined with: (i) the RSV LTR promoter (SEQ ID NO:75) and synthetic 5’ intron (SEQ ID NO 76) as well as mouse immunoglobulin heavy chain secretion leader peptide cDNA (SEQ ID NO:77) at the 5’ end and with human lgG1 constant domain cDNA (SEQ ID NO: 78) at the 3’ end for the VH fragment; and (ii) the CMV IE promoter (SEQ ID NO: 79) linked with the leader sequence from herpes virus TK gene (SEQ ID NO: 80) as well as mouse immunoglobulin light chain secretion leader peptide cDNA (SEQ ID :81) at the 5’ end and with human immunoglobulin kappa (SEQ ID: 82) or lambda (SEQ ID: 83) light chain constant domain cDNA at the 3’ end, for the V
- the reaction creates just natural joining between variable and constant domain in the human IgG 1 heavy and kappa (if the VL was amplified with kappa specific primers) or lambda (kappa if the VL was amplified with lambda specific primers) light chain, respectively.
- the final product resulting from the cloning reaction is the pQMCF IgG shuttle expression vector (SEQ ID NOs 84; 85) containing ampicillin resistance gene for selective growth of transformed E. coli and separate mammalian expression cassettes for the lgG1 heavy and light chain, respectively.
- promoters, introns, secretion signals, and constant domains used in this example are just one combination of heterologous elements that can be used to express and produce the antibodies with particular variable domain.
- a person skilled in the art would be able to find other promoters and signal elements that would work also for this purpose.
- the efficiency of the antigen-specific IgG reconstruction from VH and VL combinations was initially analyzed via the transfection of library pools.
- the DNA was transfected into Chinese hamster ovary cell line CHOEBNALT85 (Icosagen Cell Factory, Estonia) and 48-72 h later the culture supernatants were assessed by ELISA for the secretion of IgG molecules that specifically recognize S1 protein (SEQ ID NO:6) of the SARS-CoV-2, thus indicating the presence of the desired VH/VL combinations in the library.
- the library pools that showed the clearly positive signal were split to individual clones by back-transformation into competent E. coli cells and picking individual bacterial colonies - each containing single type of the plasmid with unique VH and VL combination. Then, specific S1 -binding determination by ELISA was repeated using the supernatants of CHO cells transfected with plasmid DNA preparations derived from single clones instead of library pools. Finally, the VH and VL sequences were identified by sequencing of the VH and VL insertions of the positive plasmid clones. The sequence confirmed antibody expressing clones were recombinantly expressed in CHO cells followed by affinity purification and buffer exchange procedures.
- Figure 2a-d shows analytical size exclusion chromatography results of four selected neutralizing antibody clones.
- the purified antibodies were used to determine the binding affinities of the developed anti SARS-CoV-2 Spike protein antibodies.
- A) shows binding sensograms of the four selected antibody clones and the kinetic analysis to determine the Kon, Kdiss, Kd of the developed antibodies was conducted by using Bio-Layer-Interferometry (PALL Biosciences).
- the obtained binding kinetic properties against the S1 Protein SEQ ID NO:6 are shown in B of Figure 3A-D.
- C shows binding sensograms of SARS CoV-2 antibodies of clones 23G7 and 23G7.1 . Summary table for binding parameters for these two clones is shown in part D of Figure 3A-D.
- Antibody clones 31 B9, 13A4 and 38G5 belong to a second overlapping epitope bin.
- Antibodies 23E2, 42B7 and 42H8 belong to the third overlapping epitope bin.
- As antibody 8A12 does not bind the RBD but binds the S1 domain outside of the RBD (data not shown) it is evident that in total we are able to target 4 independent epitope bins located on the S1 domain. Based on this experiment there is potential to use these antibodies as a combinational therapy option as a subset of these antibodies can bind to the S1 protein simultaneously and potentially neutralize the SARS-CoV-2 infection.
- the VH sequences were amplified from PBMCs using reverse primer (SEQ ID NO: 23) that binds to very beginning of the CH1 domain of the heavy chain constant region that is identical for all known human IgG subclasses (i.e. IgG 1 , lgG2, lgG3 , lgG4). Later, during the vector cloning process in construction of the library pools, the amplified VH sequences are artificially linked to human lgG1 constant region present in the vector backbone.
- the fragment of the antibody coding sequence including the unique c-terminal half of the VH domain and N-terminal part of the IgG heavy chain constant domain was amplified from the cDNA reaction prepared from capture reaction that resulted of each particular antibody clone.
- the PCR was using very specific forward primer (SEQ ID NO 9-22) that binds unique sequence of VH CDR2 of the antibody of interest together with reverse primer (SEQ ID NO:23) that binds to CH2 domain of all known human IgG subclasses (i.e. lgG1 , lgG2, lgG3, lgG4).
- the amplified fragments were cloned into the pJetl .2 cloning vector (available at ThermoFisher Scientific Cat. Nos. K1231 , K1232 Pub. No. MAN0012707 Rev. B.01 ) and 6-8 individual clones were sequenced for each antibody. The sequence analysis clearly revealed that all those antibodies originally belonged to lgG1 subclass in human donors.
- 23G7 VH is most close to human germline sequence IGHV1 -2*04 (97.3% identity); and 23G7 VL (lambda) is most close to human germline gene sequence IGLV2-14*01 (98.6% identity);
- 23E2 VH is most close to human germline sequence IGHV3-53*04 (99.0% identity); and 23E2 VL (kappa) is most close human germline gene sequence IGKV1 -9*01 (99.3% identity);
- 8A12 VH is most close to human germline sequence IGHV1 -24*01 (97.6% identity).
- 8A12 VL (kappa) is most close to human germline gene sequence IGKV2-24*01 (98.7% identity).
- 42B7 VH is most close to human germline sequence IGHV3-66*02 (95.9% identity); and 42B7 VL (kappa) is most close to human germline gene sequence IGKV1 -39*01 (100% identity).
- VH 23G7.1 SEQ ID NO: 72
- VL 23G7.1 SEQ ID NO: 74
- the VH 23G7.1 and VL 23G7.1 encoding DNA sequences were codon optimized resulting in the following sequence for VH (SEQ ID: 71 ) and for VL (SEQ ID: 73).
- Part C of Figure 3A-D demonstrates the comparative binding sensorgram of 23G7 IgG and 23G7.1 IgG to the trimeric Spike ectodomain and part D of Figure 3A-D demonstrates the binding parameters of both antibody variants to the trimeric Spike ectodomain. As seen from parts C and D of Figure 3A-D both antibody variants elicit identical binding properties.
- the epitopes that are recognized by the antibody in its target protein sequence can be divided to two types: linear and conformational.
- Linear epitopes consist of continuous residues on a protein sequence.
- conformational epitopes consist of residues that are discontinuous in the protein sequence but come within close proximity to form an antigenic surface on the protein's three-dimensional structure.
- linear epitopes are recognized when the target protein is completely denatured, such as heated in reducing conditions and separated in SDS- PAGE gels.
- the same S1 protein of SARS-CoV-2 that was used for initial ELISA screening and obtaining those antibodies and which is recognized by those antibodies in the ELISA assay were also tested in conditions of completely denatured protein. Briefly, 1 pg of the S1 protein was denatured in 50 pl of SDS-PAGE loading buffer with DTT as reducing agent (2% SDS; 10% glycerol; 0.002% bromphenol blue; 67.5 mM Tris HCI, pH 6.8; 100 mM DTT) by heating the sample 5 minutes at 99 °C.
- DTT as reducing agent
- the sample was loaded and run on gradient (8-16%) SDS-PAGE gel using 5 pl or 100 ng of S1 per lane. Then, the binding of the antibody was analyzed using Western blot technique with the antibodies of interest as primary antibodies (in concentrations 1 pg/ml and 5 pg/ml for 23E2, 23G7 and 8A12; and 1 pg/ml for 18B6) and anti-human IgG HRP conjugate as the secondary antibody (the same secondary antibody was successfully used for determination of the S1 binding of the same antibodies in ELISA). In this assay none of the selected antibodies showed binding to denatured S1 protein of SARS-CoV-2 (results not shown) indicating that those antibodies recognize conformational and not linear epitope in the target protein.
- a cell-based virus neutralization assay (part A of Fig. 7A-B). 4 x 10 4 Vero-E6 cells were seeded per well in 96-well plates. The cells were grown for 24 h in DMEM supplemented with 10% FBS and Pen-Strep. Antibodies were prepared in 5-fold dilutions at 7 different concentrations, starting from 0.04mg/mL in the virus growth medium (VGM) containing 0.2% BSA and Pen-Strep in
- DMEM Virus strain hCoV-19/Norway/Trondheim-E9/2020 obtained from infected patient in Norway and expanded in a BSL3 containment facility, was added to the samples to achieve a moi of 0.1 and incubated for 1 h at 37 °C. 0,1 % DMSO was added to the control wells. The Vero-E6 cells were incubated for 72 h with VGM.
- Part B of Figure 7A-B shows inhibition (neutralizing effect) of antibody clones 8A12, 23G7 and 23E2.
- 23G7 had the most potent neutralizing effect of 5.9 ng/ml, followed by 8A12 and 23E2 with IC50 values of 839.8 ng/ml and 1267 ng/ml respectively.
- Antibodies are the first line of defense for many mucosal pathogens including respiratory viruses. For both influenza and coronaviruses, the neutralizing IgA response located in the mucosal tissue has been demonstrated to be most efficacious in preventing viral infection. Antibodies have the ability to bind and block cell entry of viral pathogens, thus they are an important class of biologic therapeutic molecules that can be developed recombinantly to both protect against viral infection and neutralize an already occurred infection. Recombinant antibodies that have been cloned from convalescent humans, immunized animals or synthetic antibodies obtained from molecular display libraries can all be an excellent source for antibodies that elicit virus neutralizing properties. These antibodies administered through a systemic intravenous delivery, local delivery using nebulized antibodies for inhalation to the lower respiratory tract or as a nasal spray that can be administered to the upper respiratory tract to generate a virus neutralizing protective biolayer.
- the IgA antibody isotype is the most predominant antibody isotype located in the mucosal tissue and can form a multimeric molecule with up to a decavalent pentameric structure.
- the anti-SARS-CoV-2 antibody 23G7 described here can be expressed either as hlgG isotype antibody and it’s derivates as well as an IgA isotype molecule while retaining binding characteristics to the SARS-CoV-2 Spike protein.
- 23G7 has an extremely low IC50 value as shown here, it is a candidate to neutralize SARS-CoV-2 viral infection, and therefore it has the potential for both a preventive and therapeutic drug against COVID19 that could be administered either intravenously or locally as a nebulized molecule or a nasal spray for direct delivery to mucosal tissue in various human immunoglobulin isotype frameworks.
- nasal cavities and nasopharynx contain some of the highest viral loads in the body, and viral loads are similar in symptomatic and asymptomatic individuals. Therefore, silent spreaders may unknowingly contribute to the exponential growth of disease, as nasal secretions contain spreadable virus, and contagiousness appears to be highest before or shortly after symptom onset. For this reason, nasal applications are specifically potent to mitigate the pandemic.
- Non limiting examples of intranasal delivery of the antibodies characterized in this application, and especially antibody 23G7, but also antibodies 8A12, 23E2, 42B7, or any combinations thereof could be in form of solution sprays, saline rinses, nanogels, intranasal foams or dissolvable packings, dry nasal powder sprays, nasal ointments.
- a pharmaceutically acceptable composition suitable for patient administration will contain an effective amount of the antibody in a formulation which both retains biological activity while also promoting maximal stability during storage within an acceptable temperature range.
- the pharmaceutical compositions can also include, depending on the formulation desired, pharmaceutically acceptable diluents, pharmaceutically acceptable carriers and/or pharmaceutically acceptable excipients, or any such vehicle commonly used to formulate pharmaceutical compositions for human administration.
- the diluent is selected so as not to affect the biological activity of the combination. Examples of such diluents are distilled water, physiological phosphate-buffered saline, Ringer's solutions, dextrose solution, and Hank's solution.
- the amount of an excipient that is useful in the pharmaceutical composition or formulation of this invention is an amount that serves to uniformly distribute the antibody throughout the composition so that it can be uniformly dispersed when it is to be delivered to a subject in need thereof. It may serve to dilute the antibody to a concentration which provides the desired beneficial palliative or curative results while at the same time minimizing any adverse side effects that might occur from too high a concentration.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Virology (AREA)
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Molecular Biology (AREA)
- Medicinal Chemistry (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Oncology (AREA)
- Biophysics (AREA)
- Biochemistry (AREA)
- Immunology (AREA)
- Pulmonology (AREA)
- Communicable Diseases (AREA)
- Genetics & Genomics (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Pharmacology & Pharmacy (AREA)
- Animal Behavior & Ethology (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Peptides Or Proteins (AREA)
Abstract
Novel neutralizing SARS-CoV-2 S1 binding antibodies are provided here. The antibodies have low IC50 values and are suitable to be used separately or in combination to neutralize SARS-CoV-2 viral infection. The antibodies have potential for both a preventive and therapeutic drug against COVID19 and could be administered either intravenously or locally as a nebulized molecule or a nasal spray for direct delivery to mucosal tissue in various human immunoglobulin isotype frameworks.
Description
Title: SARS CoV-2 neutralizing antibodies
Priority
This application claims priority of US provisional application number 63/090576 filed on October 12, 2020, the contents of which are incorporated herein by reference.
Sequence listing
This application contains a sequence listing provided in computer readable format.
Field of invention
This application relates to novel antibodies and specifically to novel SARS CoV-2 neutralizing antibodies.
Background
COVID 19-pandemic is a pandemic of coronavirus disease 2019 caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). SARS-CoV-2 is a novel virus infecting human beings: first human cases of COVID-19 were identified in Wuhan, China, in December 2019.
By now the virus has infected more than 33 million people and at least 998,200 people have died. The virus has been detected in nearly every country.
The novel coronavirus has had devastating global health, as well as economic consequences and even if there are several vaccine candidates under development the treatment and prevention of the viral infection are still to be developed.
Recent studies on SARS-CoV-2 have shown that this coronavirus can bind to the alveolar pneumocytes in human lungs through their interaction with the angiotensin I and converting enzyme 2 (ACE2) surface receptor. ACE2 is an enzyme attached to the cell membranes of cells located in the lungs, arteries, heart, kidney, and intestines. As a transmembrane protein, ACE2 serves as the main entry point into cells for some coronaviruses, including SARS-CoV-2. More specifically, the binding of the spike S1 protein of SARS-CoV-2 to the enzymatic domain of ACE2 on the surface of cells results in endocytosis and translocation of both the virus and the enzyme into endosomes located within cells.
Neutralizing antibodies can block the entry of a pathogen into the cell and thus prevent the pathogen from infecting the body. As ACE2 is the main entry point for SARS-Cov-2, finding efficient neutralizing antibodies that could block the entry seems a promising approach for developing prophylactic and therapeutic means to fight against the pandemic.
Summary of the invention
Accordingly, it is an object of this invention to provide efficient neutralizing antibodies binding to the S1 -domain of the Spike protein of SARS-CoV 2 virus and competing with binding of ACE2.
According to certain aspects, the present invention provides neutralizing antibodies and antibody compositions that are suitable for administration to a subject having SARS-Cov-2 infection, or at risk of SARS-Cov-2 infection, in an amount and according to a schedule sufficient to prevent or reduce the virus infection.
According to certain aspects, the present invention provides antibodies suitable for use as a passive vaccine against the SARS-CoV-2 comprising at least one antibody of the invention and a pharmaceutically acceptable carrier.
According to certain aspects, the present invention provides antibodies suitable for use as a nasal spray to provide prophylactic treatment against SARS-CoV-2 infection.
According to certain aspects, the present invention provides an antibody-based pharmaceutical composition comprising an effective amount of one of more of the isolated SARS-CoV-2 antibodies described herein for providing a prophylactic or therapeutic treatment to reduce infection of the SARS-CoV- 2.
It is an object of this invention to provide an isolated SARS-CoV-2 S1 -domain binding antibody or fragment thereof, wherein the antibody comprises a heavy chain variable region (VH) comprising the VH CDR 1 -3 according to SEQ ID NOs 95-97, respectively, and a light chain variable region (VL) comprising the VL CDR1 -3 of SEQ ID NOs 89-91 , respectively.
According to certain aspects the isolated SARS-CoV-2 S1 -domain binding antibody or fragment thereof, wherein the antibody comprises the VH protein according to SEQ ID NO: 44 or 72 and VL protein according to SEQ ID NO: 48 or 74, respectively.
It is an object of this invention to provide an isolated SARS-CoV-2 S1 -domain binding antibody or fragment thereof, wherein the antibody comprises a heavy chain variable region VH according to SEQ ID NO: 60 and light chain variable region VL according to SEQ ID NO: 62, or VH according to SEQ ID NO: 64 and VL according to SEQ ID NO: 66, or VH according to SEQ ID NO: 68 and VL according to SEQ ID NO: 70.
It is an object of this invention to provide a composition comprising the isolated SARS-CoV-2 S1 -domain binding antibody comprising a heavy chain variable region (VH) comprising the VH CDR 1 -3 according to SEQ ID Nos: 95-97, respectively, and a light chain variable region (VL) comprising the VL CDR1 -3 of SEQ ID Nos: 89-91 , respectively.
According to certain aspects the composition additionally comprises one or more of the antibodies comprising a heavy chain variable region VH according to SEQ ID
NO: 60 and light chain variable region VL according to SEQ ID NO: 62, or VH according to SEQ ID NO: 64 and VL according to SEQ ID NO: 66, or VH according to SEQ ID NO: 68 and VL according to SEQ ID NO: 70.
According to certain aspects the composition is a pharmaceutical composition, comprising a pharmaceutically acceptable carrier.
According to certain aspects the composition is a nasal spray.
It is an object of this invention to provide a method to alleviate symptoms of COVID 19 disease, the method comprising administering to a subject an effective amount of the pharmaceutical composition.
It is an object of this invention to provide an isolated nucleic acid encoding the VH comprising the VH CDR 1 -3 according to SEQ ID NOs 95-97, respectively and /or the VL comprising the VL CDR1 -3 of SEQ ID NOs 89-91 , respectively.
It is an object of this invention to provide an isolated nucleic acid encoding the VH according to SEQ ID NO: 44 or 72 and VL according to SEQ ID NO: 48 or 74, respectively.
It is an object of this invention to provide an isolated nucleic acid encoding the VH according to SEQ ID NO: 60 and/or light chain variable region VL according to SEQ ID NO: 62, or VH according to SEQ ID NO: 64 and/or VL according to SEQ ID NO: 66, or VH according to SEQ ID NO: 68 and/or VL according to SEQ ID NO: 70.
Short description of the figures
Figure 1 illustrates the infection cycle of SARS-CoV-2. SARS-CoV-2 uses angiotensin converting enzyme 2 (ACE2) receptor to enter the host cell in which the virus then replicates. Virus inserts its RNA into the host cell, where it utilizes the host cell to produce new viral proteins and replicate the RNA genome. Viral proteins are glycosylated and transported to the host cell membrane, where the assembled
particles bud from the host cell. Neutralizing antibodies can bind to spike S1 protein and thereby prevent or impair its capability to bind to ACE-2 receptor.
Figure 2a-d shows analytical Size-Exclusion Chromatography of SARS-CoV-2 neutralizing antibody clones, a) clone 23G7; b) clone 23E2, c) clone 8A12, d) clone 42B7.
Figure 3A-D: A) shows binding sensograms of SARS CoV-2 antibodies: a) clone 8A12 sample 12#22; b) clone 23E2, sample #81 ; c) clone 23G7, sample #86, d) clone 42B7, sample #140 to the S1 -domain of SARS-Cov-2;
B) shows a summary table of binding parameters of the SARS CoV-2 antibodies of clones 8A12, 23G7, 23E2 and 42B7;
C) shows interferometry sensorgrams of SARS CoV-2 antibodies of clone 23G7 and clone 23G7.1 to the trimeric S-ectodomain protein of SARS-Cov-2
D) shows a summary table of binding parameters of SARS CoV-2 antibodies 23G7 and 23G7.1
Figure 4 shows in-tandem bio-layer interferometry binning assay of SARS-CoV-2 RBD binding antibodies indicating the RBD binding antibodies belong to three independent epitope bins.
Figure 5a-d shows ACE2 blocking assay by bio-layer interferometry, a) shows ACE2 binding of clone 8A12 antibodies; b) shows ACE2 binding of clone 23G7 antibodies; c) shows ACE 2 binding of clone 23E2 antibodies; d) shows ACE2 binding of 42B7 antibodies. All tested antibodies except 8A12 is able to block ACE2 binding.
Figure 6 shows ACE binging inhibition assay by ELISA. Samples from Clones 23G7, 23E2, 8A12 and 42B7 were tested. Various concentrations of antibodies were used to treat immobilized S-protein followed by detection with biotinylated ACE2 and Strepavidin-HRP.
Figure 7A-B: A) shows SARS CoV-2 live virus neutralization assay. VERO E6 cells were treated with hCoV-19/Norway/Trondheim-E9/2020 at moi 0.1 together with various concentrations of SARS-CoV-2 antibodies of clones 8A12, 23G7 and 23E2, respectively. Cell viability was measured 72hours post-infection. B) shows live virus assay results measuring final antibody concentration where no cytopathic effects could be detected.
Detailed description of the invention
Definitions
As used here, “antibody” is a polypeptide that specifically binds and recognizes an antigen such as S protein of SARS-Cov2 virus or an antigenic fragment thereof.
A "monoclonal antibody" is an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigenic epitope.
“Vaccine” as used here means a pharmaceutical composition that induces a prophylactic or therapeutic immune response in a subject. In some cases, the immune response is a protective immune response. Typically, a vaccine induces an antigen-specific immune response to an antigen of a pathogen, for example a viral pathogen, such as SARS-Cov-2. A vaccine may include a polynucleotide (such as a nucleic acid encoding a disclosed antigen), or a peptide or polypeptide (such as a disclosed antigen). In one example a vaccine induces an immune response that reduces the severity of the symptoms associated with SARS-Cov 2 infection and/or decreases the viral load compared to a control. In another non-limiting example, a vaccine induces an immune response that reduces and/or prevents SARS-CoV-2 infection compared to a control.
The term "disease" refers to any alternation in state of the body or of some of the organs, interrupting or disturbing the performance of the functions and/or causing symptoms such as discomfort, dysfunction, distress, or even death to the person afflicted or those in contact with a person.
As used herein the term "in need thereof" means having a disease, being diagnosed with a disease, or being in need of preventing a disease. A subject in need thereof can be a subject in need of treating or preventing a disease.
As used herein, the terms "treat," "treatment," "treating," refer to therapeutic treatments, wherein the object is to reverse, alleviate, ameliorate, inhibit, slow down or stop the progression or severity of an infectious disease such as COVID 19- disease. The term "treating" includes reducing or alleviating at least one adverse effect or symptom of an infectious disease. Treatment is generally "effective" if one or more symptoms or clinical markers are reduced. Alternatively, treatment is "effective" if the progression of a disease is reduced or halted. That is, "treatment" includes not just the improvement of symptoms or markers, but also a cessation of, or at least slowing of, progress or worsening of symptoms compared to what would be expected in the absence of treatment. Beneficial or desired clinical results include, but are not limited to, alleviation of one or more symptoms, diminishment of extent of disease, stabilized (i.e. , not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, remission (whether partial or total), and/or decreased mortality. For example, treatment is considered effective if the condition is stabilized, or the progression of COVID 19 disease is slowed or halted. The term "treatment" of a disease also includes providing relief from the symptoms or side-effects of the disease (including palliative treatment).
As used herein, the term "prevent" or "prevention" refers to stopping, hindering, and/or slowing down an infectious disease. In one embodiment, "prevent" is synonymous with "inhibit".
As used herein, the term "administering," refers to the placement of a compound as disclosed herein into a subject by a method or route which results in at least partial delivery of the agent at a desired site. Pharmaceutical compositions comprising the compounds disclosed herein can be administered by any appropriate route which results in an effective treatment in the subject.
As used herein, a "subject" means a human. The terms, "individual," "patient" and "subject" are used interchangeably herein.
The terms "increased", "increase", or "enhance" are all used herein to generally mean an increase by a statically significant amount; the terms "increased",
"increase", or "enhance", mean an increase of at least 10% as compared to a reference level, for example an increase of at least about 10%, at least about 20%, or at least about 30%, or at least about 40%, or at least about 50%, or at least about 60%, or at least about 70%, or at least about 80%, or at least about 90% or up to and including a 100% increase or any increase between 10-100% as compared to a reference level, or at least about a 2-fold, or at least about a 3-fold, or at least about a 4-fold, or at least about a 5-fold or at least about a 10-fold increase, or any increase between 2-fold and 10-fold or greater as compared to a reference level.
The terms "decrease", "reduce", "reduction", or "inhibit" are all used herein generally to mean a decrease by a statistically significant amount. For example, "decrease", "reduce", "reduction", or "inhibit" means a decrease by at least 10% as compared to a reference level, for example a decrease by at least about 20%, or at least about 30%, or at least about 40%, or at least about 50%, or at least about 60%, or at least about 70%, or at least about 80%, or at least about 90% or up to and including a 100% decrease (e.g. absent level or non-detectable level as compared to a reference level), or any decrease between 10-100% as compared to a reference level. In the context of a marker or symptom is meant a statistically significant decrease in such level. The decrease can be, for example, at least 10%, at least 20%, at least 30%, at least 40% or more, and is preferably down to a level accepted as within the range of normal for an individual without a given disease.
As used here, the term "pharmaceutically acceptable" refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
As used here, the term "pharmaceutically acceptable carrier" means a pharmaceutically-acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, manufacturing aid (e.g., lubricant, talc magnesium, calcium or zinc stearate, or steric acid), or solvent encapsulating material necessary or used in formulating an active ingredient or agent for delivery to a subject. Each carrier must be "acceptable" in the sense of being compatible with the other
ingredients of the formulation and not injurious to the patient.
As used herein, the term "in combination" refers to the use of more than one prophylactic and/or therapeutic agent simultaneously or sequentially and in a manner that their respective effects are additive or synergistic.
“Composition” as used herein may be a single or a combination of antibodies disclosed herein, which can be the same or different, in order to prophylactically or therapeutically treat the progression SARS Cov-2 infection after administration. Such combinations can be selected according to the desired immunity or effect.
The antibody-based pharmaceutical composition of the present invention may be formulated by any number of strategies known in the art (e.g., see McGoff and Scher, 2000, Solution Formulation of Proteins/Peptides: In McNally, E. J., ed. Protein Formulation and Delivery. New York, N.Y.: Marcel Dekker; pp. 139-158; Akers and Defilippis, 2000, Peptides and Proteins as Parenteral Solutions. In: Pharmaceutical Formulation Development of Peptides and Proteins. Philadelphia, Pa.: Talyor and Francis; pp. 145-177; Akers, et al., 2002, Pharm. Biotechnol. 14:47-127).
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) emerged at the end of 2019 and was shortly thereafter sequenced. SARS-CoV-2 appears to have originated in bats from which closely related viruses and viral sequences have been identified. SARS-CoV-2 belongs to the sarbecovirus subgenus and is closely related to SARS-CoV which was responsible for an epidemic in 2002-2003 resulting in 8,098 cases and 774 fatalities worldwide. The lack of pre-existing immunity to SARS-CoV- 2 due to its divergence from the circulating endemic coronaviruses, and its efficient human-to-human transmission has resulted in the ongoing COVID-19 pandemic which has already caused more than 33 million infections and over million fatalities as of late-September 2020. SARS-CoV-2 infection starts upon attachment of the viral trimeric transmembrane spike (S) glycoprotein via a receptor-binding domain (RBD) to angiotensin-converting enzyme 2 glycoprotein (ACE2), leading to membrane fusion and entry into host cells c.f. Figure 1 . As for other coronaviruses, SARS-CoV- 2 spike protein is the main target of neutralizing antibodies (Abs) and a focus of vaccine design and therapeutic targeting strategies. Although vaccine development programs are progressing rapidly, large-scale manufacturing and administration to a
large enough population for achieving community protection will probably take several months. Prophylactic and/or therapeutic anti-viral drugs could allow overcome the gap before safe and efficient vaccines become widely available and will continue to be useful in unvaccinated individuals or those who respond poorly to vaccination. Unlike controversial FDA-approved convalescent plasma, a treatment that takes whole plasma from a recovered person, and infuses it into another patient with the disease, monoclonal antibody therapy uses a much more targeted approach, specifically neutralizing SARS-CoV-2 infectivity. Therefore, SARS-CoV-2- targeting antibodies are really valuable as they are supposed to be an important bridge to a vaccine and a hedge in the event vaccines are delayed or work inefficiently. Similar to vaccines, antibody treatments are also being developed to prevent SARS-CoV-2 infection, particularly in people who are at high risk and who might have been exposed to the virus through close contact with an already infected person. These antibodies are designed to neutralize the virus in patients who are sick or offer short-term immunity to people who have been exposed. Passive immunization with neutralizing anti-SARS-CoV-2 antibody could be especially valuable for certain populations that are suffering the most: the elderly, the immunocompromised, patients in nursing homes and long-term care facilities. Signs and symptoms of COVID-19 may appear two to 14 days after exposure. This time after exposure and before having symptoms is called the incubation period. Common signs and symptoms can include fever, cough, tiredness. Other symptoms can include shortness of breath or difficulty breathing, muscle aches, chills, sore throat, runny nose, headache, chest pain. The severity of COVID-19 symptoms can range from very mild to severe. Some people may have only a few symptoms, and some people may have no symptoms at all. Some people may experience worsened symptoms, such as worsened shortness of breath and pneumonia, about a week after symptoms start.
This invention provides novel neutralizing anti-SARS-CoV-2 antibodies. The invention is now described with examples and references to the figures.
Human donors and peripheral blood mononuclear cell (PBMC) samples used for obtaining of the antibodies recognizing the S1 protein of SARS-CoV-2.
Human sample collection and further processing were approved by Research Ethics Committee of the University of Tartu (No. 305/T-1 , 07.04.2020). Monoclonal antibodies recognizing S1 protein ( SEQ ID NO:6) of the SARS-CoV-2 virus or, more specifically, receptor binding domain (RBD) (SEQ ID NO: 2 or 4) of the S1 protein were isolated from human subjects diagnosed with COVID-19 by detection of the SARS-CoV-2 virus RNA in nasal swab sample by RT-PCR. Peripheral blood mononuclear cells (PBMCs) were collected 2-4 weeks after diagnosis and at this time the donors were recovered from the infection and had no acute COVID-19 infection symptoms.
Venous blood samples were collected and PBMCs were separated into BD Vacutainer® CPT™ Cell Preparation Tubes containing the cell separation medium (BD Biosciences, Cat No.362753) according to manufacturer’s instructions. The collected PBMCs were stored under liquid nitrogen up to further processing for the isolation of the monoclonal antibodies.
Isolation of S1/RBD binding human monoclonal antibodies using Hybrifree technology
Monoclonal antibodies recognizing SARS-Cov-2 S1 protein were isolated using Hybrifree technology (Kivi G, et al., BMC Biotechnol. 2016; 16:2 incorporated herein by reference). Briefly, the frozen PBMCs was thawed, washed and collected into 10 ml of RPMI1640 supplemented with penicillin/streptomycin and 10 % of heat inactivated fetal bovine serum. Then the cells were seeded into a 100 mm cell culture dish and incubated ~1 h at 37 °C in an 8 % CO2 atmosphere. Then, free- floating cells (the fraction enriched for B-cells and separated from plastic-adherent cells, e.g. macrophages) were collected, viable cell count was determined and the cells were transferred into the capture medium (RPMI1640 supplemented with 0.5 % BSA and 0.1 % NaN3).
For capture or panning of the B-cells that express SARS-CoV-2 S1 protein specific antibodies on their surface, MaxiSorp™ surface 96-wells (Thermo Fisher Scientific, US) were coated with the recombinant S1 subunit (SEQ ID NO:6) of the spike protein (SEQ ID NO:8) or RBD domain (SEQ ID NO:4) of the S1 and blocked for 1 h with 2 % BSA in PBS. One hundred microliters of cell suspension containing 1 x 104
live cells in capture medium were loaded into a single well. The plate was centrifuged (200 x g, 5 min) and incubated for 45-60 min. The medium was discarded, and loosely attached cells were removed by washing 4-5 times with PBS. Finally, the plastic-bound cells were lysed and subjected to total RNA isolation and cDNA synthesis using oligo-T primer. The cDNA synthesis was conducted using SuperScript™ IV First-Strand Synthesis System (Thermo Fischer Scientific Catalog number: 18091050) and oligo d(T)2o primer supplied with the kit.
The cDNAs of antibody VH and VL regions were amplified using cocktails of forward primers that bind to FR1 regions of VH or VL kappa and VLJambda light chain coding sequences and were designed to maximally cover the variability of the human VH and VL sequences. In each reaction, single reverse primer was used that binds to the beginning of the constant region encoding sequence of the human IgG heavy chains, kappa light chain or lambda light chain, respectively.
The VH forward primers (5’-3’) were according to SEQ ID NO:9 - SEQ ID NO: 22.
The VH reverse primer (5’-3’) was according to SEQ ID NO:23.
The VL kappa forward primers (5’- 3’) were according to SEQ ID NO 24-33.
The VL kappa reverse primer was according to SEQ ID NO:34.
The VL lambda forward primers (5’-3’) were according to SEQ ID NO: 35-41 and SEQ ID NO:98.
The VL lambda reverse primer was according to SEQ ID NO: 42.
Using the ligase independent cloning strategy, VH and VL PCR products retrieved from the same capture reaction were both exactly joined with: (i) the RSV LTR promoter (SEQ ID NO:75) and synthetic 5’ intron (SEQ ID NO 76) as well as mouse immunoglobulin heavy chain secretion leader peptide cDNA (SEQ ID NO:77) at the 5’ end and with human lgG1 constant domain cDNA (SEQ ID NO: 78) at the 3’ end for the VH fragment; and (ii) the CMV IE promoter (SEQ ID NO: 79) linked with the leader sequence from herpes virus TK gene (SEQ ID NO: 80) as well as mouse immunoglobulin light chain secretion leader peptide cDNA (SEQ ID :81) at the 5’ end and with human immunoglobulin kappa (SEQ ID: 82) or lambda (SEQ ID: 83) light chain constant domain cDNA at the 3’ end, for the VL fragment. The reaction creates
just natural joining between variable and constant domain in the human IgG 1 heavy and kappa (if the VL was amplified with kappa specific primers) or lambda (kappa if the VL was amplified with lambda specific primers) light chain, respectively. The final product resulting from the cloning reaction is the pQMCF IgG shuttle expression vector (SEQ ID NOs 84; 85) containing ampicillin resistance gene for selective growth of transformed E. coli and separate mammalian expression cassettes for the lgG1 heavy and light chain, respectively. It is to be understood that the promoters, introns, secretion signals, and constant domains used in this example are just one combination of heterologous elements that can be used to express and produce the antibodies with particular variable domain. A person skilled in the art would be able to find other promoters and signal elements that would work also for this purpose.
Transformation of the cloning reactions to competent E. coli cells and the bacterial culture was propagated in liquid media. Then purification of the plasmid DNA products from the propagated bacteria resulted library pools of the antibody expressing vectors. In principle, such library pools are VH/VL combinatory libraries of limited numbers of VH and VL sequences retrieved from the same capture reaction and linked pairwise in the single expression vector molecule.
The efficiency of the antigen-specific IgG reconstruction from VH and VL combinations was initially analyzed via the transfection of library pools. The DNA was transfected into Chinese hamster ovary cell line CHOEBNALT85 (Icosagen Cell Factory, Estonia) and 48-72 h later the culture supernatants were assessed by ELISA for the secretion of IgG molecules that specifically recognize S1 protein (SEQ ID NO:6) of the SARS-CoV-2, thus indicating the presence of the desired VH/VL combinations in the library.
Next, the library pools that showed the clearly positive signal were split to individual clones by back-transformation into competent E. coli cells and picking individual bacterial colonies - each containing single type of the plasmid with unique VH and VL combination. Then, specific S1 -binding determination by ELISA was repeated using the supernatants of CHO cells transfected with plasmid DNA preparations derived from single clones instead of library pools. Finally, the VH and VL sequences were identified by sequencing of the VH and VL insertions of the positive plasmid clones. The sequence confirmed antibody expressing clones were recombinantly
expressed in CHO cells followed by affinity purification and buffer exchange procedures. Figure 2a-d shows analytical size exclusion chromatography results of four selected neutralizing antibody clones. The purified antibodies were used to determine the binding affinities of the developed anti SARS-CoV-2 Spike protein antibodies. In Figure 3A-D, A) shows binding sensograms of the four selected antibody clones and the kinetic analysis to determine the Kon, Kdiss, Kd of the developed antibodies was conducted by using Bio-Layer-Interferometry (PALL Biosciences). The obtained binding kinetic properties against the S1 Protein SEQ ID NO:6 are shown in B of Figure 3A-D. In figure 3A-D, C shows binding sensograms of SARS CoV-2 antibodies of clones 23G7 and 23G7.1 . Summary table for binding parameters for these two clones is shown in part D of Figure 3A-D.
Identifying ACE2 blocking clones by bio-layer interferometry and ELISA based competition assays.
As antibodies capable of ACE2 blocking have been characterized to elicit virus neutralizing capacity (see Figure 1 ), we performed competition binding assays by bio-layer interferometry (BLI) and ELISA. For BLI assays we loaded the sensor tip with the SARS-CoV-2 specific antibody from the above antibody clones, followed by a first binding event of S1 -domain protein. This was followed by dipping the sensor in ACE2 containing buffer for a determining a secondary binding event. As demonstrated in figure 4 antibody clones 23G7, 23E2 and 42B7 were able to block the secondary binding event, indicating that they are competing with ACE2 for binding with the RBD. 8A12 was not able to block ACE2 binding.
To confirm these results, we performed an ELISA based competition assay (Figure 5a-d, Figure 6). S-protein (SEQ ID NO:8) was coated at 4ug/ml on a MaxiSorp high- capacity binding ELISA plate (Thermo Fisher Scientific, US). S-protein was incubated with antibodies from various clones at various concentrations of the antibodies, followed by washes and incubation with biotinylated ACE2-Fc and detection by Strepavidin-HRP. Concentration dependent blocking of ACE2-Fc was determined for the antibodies that was in correlation with data obtained with BLI
assays. Antibody 8A12 binds to the S1 domain but does not bind to the RBD and does not block ACE2-Fc binding at any concentration.
Epitope binning assays
In order to determine if the obtained anti-S protein antibodies enable simultaneous binding to the S-protein we performed an in-tandem protocol binning experiment by BLI (Fig. 4). Seven clones of antibodies were selected for the assays. The S1 protein was captured to the sensor followed by capture of the 1 st mAb. Subsequently the sensor was dipped in a buffer containing the second competing antibody. A secondary binding event with the competing mAb indicates non-competing binding of the tested antibody pair. As evidenced in Figure 4, the RBD specific antibodies belong to 3 independent epitope bins. Antibody 23G7 belongs to an independent epitope bin by blocking of all other tested RBD specific antibodies. Antibody clones 31 B9, 13A4 and 38G5 belong to a second overlapping epitope bin. Antibodies 23E2, 42B7 and 42H8 belong to the third overlapping epitope bin. As antibody 8A12 does not bind the RBD but binds the S1 domain outside of the RBD (data not shown) it is evident that in total we are able to target 4 independent epitope bins located on the S1 domain. Based on this experiment there is potential to use these antibodies as a combinational therapy option as a subset of these antibodies can bind to the S1 protein simultaneously and potentially neutralize the SARS-CoV-2 infection.
IgG subtype determination of the selected antibodies
According to the method used to obtain the antibodies as described above, the VH sequences were amplified from PBMCs using reverse primer (SEQ ID NO: 23) that binds to very beginning of the CH1 domain of the heavy chain constant region that is identical for all known human IgG subclasses (i.e. IgG 1 , lgG2, lgG3 , lgG4). Later, during the vector cloning process in construction of the library pools, the amplified VH sequences are artificially linked to human lgG1 constant region present in the vector backbone. Thus, according to the method and the used vector, all antibodies obtained were initially produced and screened as human IgG 1 subtype antibodies and the initial sequence analysis of the VH region could not reveal the original IgG
subtype (determined by heavy chain constant region) of the discovered antibody into which it belonged in human donor. However, it is well known that each subclass has a unique profile with respect to antigen binding, immune complex formation, complement activation, triggering of effector cells, half-life, and placental transport. Thus, we determined the original subclass belongings of the selected antibodies 23E2, 23G7 and 18B6. For this, the fragment of the antibody coding sequence including the unique c-terminal half of the VH domain and N-terminal part of the IgG heavy chain constant domain was amplified from the cDNA reaction prepared from capture reaction that resulted of each particular antibody clone. To amplify the coding sequence of the selected antibody linked with any IgG constant region, the PCR was using very specific forward primer (SEQ ID NO 9-22) that binds unique sequence of VH CDR2 of the antibody of interest together with reverse primer (SEQ ID NO:23) that binds to CH2 domain of all known human IgG subclasses (i.e. lgG1 , lgG2, lgG3, lgG4). The sequence differences in the region from the beginning of the constant domain to the reverse primer binding site in CH2 domain are sufficient for unambiguous determination of the IgG subclass (i.e. IgG 1 , lgG2, lgG3, lgG4) of the particular antibody. On the other hand, the identity of amplified unique sequences from CDR2 to the end of the VH confirms that the VH-CH1-CH2 fragment here was amplified from the very same cDNA that resulted the initially obtaining clones 23E2, 23G7 or 18B6.
The amplified fragments were cloned into the pJetl .2 cloning vector (available at ThermoFisher Scientific Cat. Nos. K1231 , K1232 Pub. No. MAN0012707 Rev. B.01 ) and 6-8 individual clones were sequenced for each antibody. The sequence analysis clearly revealed that all those antibodies originally belonged to lgG1 subclass in human donors.
Sequence analysis of the selected monoclonal antibodies
The primary sequence analysis of the selected antibodies was conducted using IgBLAST tool (US National Library of Medicine)
https://www.ncbi. nlm.nih.gov/igblast/). The analysis using VH and VL cDNA data revealed that:
(i) 23G7 VH is most close to human germline sequence IGHV1 -2*04 (97.3% identity); and 23G7 VL (lambda) is most close to human germline gene sequence IGLV2-14*01 (98.6% identity);
(ii) 23E2 VH is most close to human germline sequence IGHV3-53*04 (99.0% identity); and 23E2 VL (kappa) is most close human germline gene sequence IGKV1 -9*01 (99.3% identity);
(iii) 8A12 VH is most close to human germline sequence IGHV1 -24*01 (97.6% identity).; and 8A12 VL (kappa) is most close to human germline gene sequence IGKV2-24*01 (98.7% identity).
(iv) 42B7 VH is most close to human germline sequence IGHV3-66*02 (95.9% identity); and 42B7 VL (kappa) is most close to human germline gene sequence IGKV1 -39*01 (100% identity).
The CDR sequence boundaries were determined as published previously. (Ehrenmann F., Kaas Q, and Lefranc M.P. Nucleic Acids res., 38: D 301 -D307 (2010). PMID: 19900967; and Ehrenmann, F., Lefranc, M-P- Cold Spring Harbor Protoc., 6:737-749 (2011 ) PMID: 21632775
As the amplification of 23G7 clone VH and VL sequences using the established primer sequence cocktail generated a 5’ FR1 region mutation when comparing to the parental germline sequence, we also generated the corrected FR1 sequence variant of VH and VL of 23G7 identified as VH 23G7.1 (SEQ ID NO: 72) and VL 23G7.1 (SEQ ID NO: 74). The VH 23G7.1 and VL 23G7.1 encoding DNA sequences were codon optimized resulting in the following sequence for VH (SEQ ID: 71 ) and for VL (SEQ ID: 73). Part C of Figure 3A-D demonstrates the comparative binding sensorgram of 23G7 IgG and 23G7.1 IgG to the trimeric Spike ectodomain and part D of Figure 3A-D demonstrates the binding parameters of both antibody variants to the trimeric Spike ectodomain. As seen from parts C and D of Figure 3A-D both antibody variants elicit identical binding properties.
Determination of the epitope type of the selected antibodies
The epitopes that are recognized by the antibody in its target protein sequence can be divided to two types: linear and conformational. Linear epitopes consist of continuous residues on a protein sequence. In contrast, conformational epitopes consist of residues that are discontinuous in the protein sequence but come within close proximity to form an antigenic surface on the protein's three-dimensional structure. Thus, only linear epitopes are recognized when the target protein is completely denatured, such as heated in reducing conditions and separated in SDS- PAGE gels.
To determine the type of epitope for the selected antibodies 23E2, 23G7, 8A12, 18B6, the same S1 protein of SARS-CoV-2 that was used for initial ELISA screening and obtaining those antibodies and which is recognized by those antibodies in the ELISA assay were also tested in conditions of completely denatured protein. Briefly, 1 pg of the S1 protein was denatured in 50 pl of SDS-PAGE loading buffer with DTT as reducing agent (2% SDS; 10% glycerol; 0.002% bromphenol blue; 67.5 mM Tris HCI, pH 6.8; 100 mM DTT) by heating the sample 5 minutes at 99 °C. Then the sample was loaded and run on gradient (8-16%) SDS-PAGE gel using 5 pl or 100 ng of S1 per lane. Then, the binding of the antibody was analyzed using Western blot technique with the antibodies of interest as primary antibodies (in concentrations 1 pg/ml and 5 pg/ml for 23E2, 23G7 and 8A12; and 1 pg/ml for 18B6) and anti-human IgG HRP conjugate as the secondary antibody (the same secondary antibody was successfully used for determination of the S1 binding of the same antibodies in ELISA). In this assay none of the selected antibodies showed binding to denatured S1 protein of SARS-CoV-2 (results not shown) indicating that those antibodies recognize conformational and not linear epitope in the target protein.
Cell based virus neutralization assays
In order to determine the ability of the developed antibodies to neutralize viral infection we performed a cell-based virus neutralization assay (part A of Fig. 7A-B). 4 x 104 Vero-E6 cells were seeded per well in 96-well plates. The cells were grown for 24 h in DMEM supplemented with 10% FBS and Pen-Strep. Antibodies were prepared in 5-fold dilutions at 7 different concentrations, starting from 0.04mg/mL in
the virus growth medium (VGM) containing 0.2% BSA and Pen-Strep in
DMEM. Virus strain hCoV-19/Norway/Trondheim-E9/2020 obtained from infected patient in Norway and expanded in a BSL3 containment facility, was added to the samples to achieve a moi of 0.1 and incubated for 1 h at 37 °C. 0,1 % DMSO was added to the control wells. The Vero-E6 cells were incubated for 72 h with VGM.
After the incubation period, the medium was removed, and a CellTiter-Glo assay was performed to measure viability.
Part B of Figure 7A-B shows inhibition (neutralizing effect) of antibody clones 8A12, 23G7 and 23E2.
From the obtained cell viability readouts after treatment at various concentrations of the antibodies we were able to determine IC50 values for the developed antibodies. Table 5 below show the IC50 values for each of the clones:
23G7 had the most potent neutralizing effect of 5.9 ng/ml, followed by 8A12 and 23E2 with IC50 values of 839.8 ng/ml and 1267 ng/ml respectively.
For antibody 42B7 and the IgA isotype antibodies of 23G7.1 we performed a live virus neutralization assay where we identified final antibody concentrations in which treating the virus we could not see any cytopathic effects caused by viral infection, indicating a minimal antibody neutralization titer. In detail, the SARS-CoV-2 (Estonian isolate 3542) was diluted in DMEM/0.2% BSA/Pen-Strep 50 ul/well and added to the serially diluted antibodies. Mixtures were incubated for 1 h at 37C. Suspension of Vero E6 cells was added to the wells of a 96 well tissue culture plate in triplicates (4x 10e4 cells/well) at a final volume 100 ul/well. The virus treated cells were incubated at 37C, 5% CO2 for 5 days. Results of neutralization assay was evaluated microscopically by detecting appearance of cytopathic effect (CPE). The first dilution, where CPE was absent was determined as the neutralizing titer. The results of the neutralization assays is demonstrated in part B of Figure 7A-B.
Examples of application
Antibodies are the first line of defense for many mucosal pathogens including respiratory viruses. For both influenza and coronaviruses, the neutralizing IgA response located in the mucosal tissue has been demonstrated to be most efficacious in preventing viral infection. Antibodies have the ability to bind and block cell entry of viral pathogens, thus they are an important class of biologic therapeutic molecules that can be developed recombinantly to both protect against viral infection and neutralize an already occurred infection. Recombinant antibodies that have been cloned from convalescent humans, immunized animals or synthetic antibodies obtained from molecular display libraries can all be an excellent source for antibodies that elicit virus neutralizing properties. These antibodies administered through a systemic intravenous delivery, local delivery using nebulized antibodies for inhalation to the lower respiratory tract or as a nasal spray that can be administered to the upper respiratory tract to generate a virus neutralizing protective biolayer.
The IgA antibody isotype is the most predominant antibody isotype located in the mucosal tissue and can form a multimeric molecule with up to a decavalent pentameric structure. The anti-SARS-CoV-2 antibody 23G7 described here can be expressed either as hlgG isotype antibody and it’s derivates as well as an IgA isotype molecule while retaining binding characteristics to the SARS-CoV-2 Spike protein. As 23G7 has an extremely low IC50 value as shown here, it is a candidate to neutralize SARS-CoV-2 viral infection, and therefore it has the potential for both a preventive and therapeutic drug against COVID19 that could be administered either intravenously or locally as a nebulized molecule or a nasal spray for direct delivery to mucosal tissue in various human immunoglobulin isotype frameworks.
The nasal cavity and nasopharynx contain some of the highest viral loads in the body, and viral loads are similar in symptomatic and asymptomatic individuals. Therefore, silent spreaders may unknowingly contribute to the exponential growth of disease, as nasal secretions contain spreadable virus, and contagiousness appears to be highest before or shortly after symptom onset. For this reason, nasal applications are specifically potent to mitigate the pandemic. Non limiting examples
of intranasal delivery of the antibodies characterized in this application, and especially antibody 23G7, but also antibodies 8A12, 23E2, 42B7, or any combinations thereof could be in form of solution sprays, saline rinses, nanogels, intranasal foams or dissolvable packings, dry nasal powder sprays, nasal ointments.
A pharmaceutically acceptable composition suitable for patient administration will contain an effective amount of the antibody in a formulation which both retains biological activity while also promoting maximal stability during storage within an acceptable temperature range. The pharmaceutical compositions can also include, depending on the formulation desired, pharmaceutically acceptable diluents, pharmaceutically acceptable carriers and/or pharmaceutically acceptable excipients, or any such vehicle commonly used to formulate pharmaceutical compositions for human administration. The diluent is selected so as not to affect the biological activity of the combination. Examples of such diluents are distilled water, physiological phosphate-buffered saline, Ringer's solutions, dextrose solution, and Hank's solution. The amount of an excipient that is useful in the pharmaceutical composition or formulation of this invention is an amount that serves to uniformly distribute the antibody throughout the composition so that it can be uniformly dispersed when it is to be delivered to a subject in need thereof. It may serve to dilute the antibody to a concentration which provides the desired beneficial palliative or curative results while at the same time minimizing any adverse side effects that might occur from too high a concentration.
Claims
1 . An isolated SARS-CoV-2 S1 domain RBD binding antibody or fragment thereof, wherein the antibody comprises a heavy chain variable region (VH) comprising the VH CDR 1 -3 according to SEQ ID Nos: 95-97, respectively, and a light chain variable region (VL) comprising the VL CDR1 -3 of SEQ ID Nos: 89-91 , respectively.
2. The isolated SARS-CoV-2 S1 RBD binding antibody or fragment thereof according to claim 1 , wherein the antibody comprises the VH according to SEQ ID NO: 44 or 72 and VL according to SEQ ID NO: 48 or 74, respectively.
3. The isolated SARS-CoV-2 S1 RBD binding antibody or fragment thereof according to claim 1 , wherein the antibody has a IC50 live virus neutralization values below 1300, more preferably below 900 and most preferably below 10 ng/ml.
4. An isolated SARS-CoV-2 S1 domain binding antibody or fragment thereof, wherein the antibody comprises a heavy chain variable region VH according to SEQ ID NO: 60 and light chain variable region VL according to SEQ ID NO: 62, or VH according to SEQ ID NO: 64 and VL according to SEQ ID NO: 66 or VH according to SEQ ID NO: 68 and VL according to SEQ ID NO: 70.
5. A composition comprising the isolated SARS-CoV-2 RBD binding antibody according to any of claims 1 -3.
6. A composition comprising the isolated SARS-CoV-2 SI domain binding antibody of claim 4.
7. The composition of claim 5, wherein the composition additionally comprises one or more of the antibodies of claim 4.
8. The composition of any of claims 5 to 7, wherein the composition is a pharmaceutical composition, comprising a pharmaceutically acceptable carrier.
9. The composition of claim 8, wherein the composition is administered intravenously or locally as a nebulized molecule or a nasal spray.
10. A method to alleviate symptoms of COVID 19 disease, the method comprising administering to a subject an effective amount of the composition of any of claims 5-9.
11 . An isolated nucleic acid encoding any of the VHs and /or the VLs of claim 1 or 2.
12. An isolated nucleic acid encoding any of the VHs and /of the VLs of claim 4.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US202063090576P | 2020-10-12 | 2020-10-12 | |
US63/090,576 | 2020-10-12 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2022079606A1 true WO2022079606A1 (en) | 2022-04-21 |
Family
ID=78302844
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/IB2021/059363 WO2022079606A1 (en) | 2020-10-12 | 2021-10-12 | Sars cov-2 neutralizing antibodies |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2022079606A1 (en) |
Citations (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN111620946A (en) * | 2020-05-09 | 2020-09-04 | 江苏省疾病预防控制中心(江苏省公共卫生研究院) | Isolated novel coronavirus monoclonal antibodies or antigen binding portions thereof |
US10787501B1 (en) * | 2020-04-02 | 2020-09-29 | Regeneron Pharmaceuticals, Inc. | Anti-SARS-CoV-2-spike glycoprotein antibodies and antigen-binding fragments |
-
2021
- 2021-10-12 WO PCT/IB2021/059363 patent/WO2022079606A1/en active Application Filing
Patent Citations (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US10787501B1 (en) * | 2020-04-02 | 2020-09-29 | Regeneron Pharmaceuticals, Inc. | Anti-SARS-CoV-2-spike glycoprotein antibodies and antigen-binding fragments |
CN111620946A (en) * | 2020-05-09 | 2020-09-04 | 江苏省疾病预防控制中心(江苏省公共卫生研究院) | Isolated novel coronavirus monoclonal antibodies or antigen binding portions thereof |
Non-Patent Citations (9)
Title |
---|
AKERS ET AL., PHARM. BIOTECHNOL., vol. 14, 2002, pages 47 - 127 |
AKERSDEFILIPPIS: "Pharmaceutical Formulation Development of Peptides and Proteins", 2000, TALYOR AND FRANCIS, article "Peptides and Proteins as Parenteral Solutions", pages: 145 - 177 |
BROUWER PHILIP J M: "Supplementary table S1 Potent neutralizing antibodies from COVID-19 patients define multiple targets of vulnerability", 15 June 2020 (2020-06-15), XP055859677, Retrieved from the Internet <URL:https://www.science.org/doi/10.1126/science.abc5902> [retrieved on 20211110], DOI: 10.1126/science.abc5902 * |
CAO YUNLONG ET AL: "Potent Neutralizing Antibodies against SARS-CoV-2 Identified by High-Throughput Single-Cell Sequencing of Convalescent Patients' B Cells", CELL, ELSEVIER, AMSTERDAM NL, vol. 182, no. 1, 9 July 2020 (2020-07-09), pages 73 - 84, 16, XP086211425, ISSN: 0092-8674, [retrieved on 20200518], DOI: 10.1016/J.CELL.2020.05.025 * |
EHRENMANN F.KAAS QLEFRANC M.P., NUCLEIC ACIDS RES, vol. 38, 2010, pages D301 - D307 |
EHRENMANN, F.LEFRANC, M-P, COLD SPRING HARBOR PROTOC, vol. 6, 2011, pages 737 - 749 |
KIVI G ET AL., BMC BIOTECHNOL, vol. 16, 2016, pages 2 |
MCGOFFSCHER: "Protein Formulation and Delivery", 2000, MARCEL DEKKER, article "Solution Formulation of Proteins/Peptides", pages: 139 - 158 |
PHILIP J. M. BROUWER ET AL: "Potent neutralizing antibodies from COVID-19 patients define multiple targets of vulnerability", SCIENCE, vol. 369, no. 6504, 7 August 2020 (2020-08-07), US, pages 643 - 650, XP055737200, ISSN: 0036-8075, DOI: 10.1126/science.abc5902 * |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
RU2526517C2 (en) | Respiratory syncytial virus (rsv) anti-g protein antibodies | |
US11730813B2 (en) | Broadly neutralizing antibodies directed against the rabies virus glycoprotein and uses thereof | |
EA034767B1 (en) | Human antibodies to respiratory syncytial virus f protein and methods of use thereof | |
US20210388065A1 (en) | Antibodies to sars-coronavirus (covid-19) s1 spike protein | |
SA96170384B1 (en) | Methods of preparation and therapeutic uses (RSV) Human neutralizing monoclonal antibodies with high affinity for the F-protein of respiratory syncytial virus | |
CA3202566A1 (en) | Neutralizing monoclonal antibodies against covid-19 | |
US10017562B2 (en) | RSV G protein specific antibodies | |
US20070292390A1 (en) | Broadly Cross-Reactive Hiv-1 Neutralizing Human Monoclonal Antibodies | |
US12054536B2 (en) | Antibodies to Andes hantavirus, and methods for using same | |
JP2020097569A (en) | Treatment and functional cure of hiv infection with anti-cd4 monoclonal antibodies that mediate competitive hiv entry inhibition | |
Ringel et al. | Optimization of the EC26-2A4 epitope in the gp41 membrane proximal external region targeted by neutralizing antibodies from an elite controller | |
US20230159626A1 (en) | Epstein-barr virus antibodies and uses thereof | |
US20220340658A1 (en) | NEUTRALIZING ANTI-SARS-CoV-2 ANTIBODIES AND USE THEREOF | |
WO2022079606A1 (en) | Sars cov-2 neutralizing antibodies | |
WO2022127739A1 (en) | Antigen-binding protein specifically binding to sars-cov-2 | |
TW202306980A (en) | Anti-respiratory syncytial virus antibody and application thereof | |
WO2023131262A1 (en) | Antigen-binding protein specifically bound to sars-cov-2 | |
US20240218056A1 (en) | Nucleocapsid-specific antibodies and methods for the treatment and prevention of sars-cov-2 infection therewith | |
CN112574297B (en) | Monoclonal antibody against neuraminidase and application thereof | |
RU2644202C2 (en) | Single-domain antibodies to ebola virus gp protein for immunotherapy of ebola fever | |
Lebedin et al. | SARS-CoV-2 neutralization and protection of hamsters via nasal administration of a humanized neutralizing antibody | |
TW202229333A (en) | Coronavirus-binding molecules and methods of use thereof |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 21794988 Country of ref document: EP Kind code of ref document: A1 |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
122 | Ep: pct application non-entry in european phase |
Ref document number: 21794988 Country of ref document: EP Kind code of ref document: A1 |