WO2022232557A1 - Compounds specific to coronavirus s protein and uses thereof - Google Patents
Compounds specific to coronavirus s protein and uses thereof Download PDFInfo
- Publication number
- WO2022232557A1 WO2022232557A1 PCT/US2022/027004 US2022027004W WO2022232557A1 WO 2022232557 A1 WO2022232557 A1 WO 2022232557A1 US 2022027004 W US2022027004 W US 2022027004W WO 2022232557 A1 WO2022232557 A1 WO 2022232557A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- day
- days
- antibody
- antigen
- subject
- Prior art date
Links
- 241000711573 Coronaviridae Species 0.000 title claims abstract description 141
- 108090000623 proteins and genes Proteins 0.000 title description 106
- 102000004169 proteins and genes Human genes 0.000 title description 64
- 150000001875 compounds Chemical class 0.000 title description 9
- 230000027455 binding Effects 0.000 claims abstract description 500
- 239000000427 antigen Substances 0.000 claims abstract description 434
- 108091007433 antigens Proteins 0.000 claims abstract description 434
- 102000036639 antigens Human genes 0.000 claims abstract description 434
- 239000012634 fragment Substances 0.000 claims abstract description 305
- 238000000034 method Methods 0.000 claims abstract description 201
- 241000315672 SARS coronavirus Species 0.000 claims abstract description 69
- 241001678559 COVID-19 virus Species 0.000 claims abstract description 61
- 230000003472 neutralizing effect Effects 0.000 claims abstract description 28
- 238000006386 neutralization reaction Methods 0.000 claims abstract description 25
- 210000002966 serum Anatomy 0.000 claims description 168
- 125000003275 alpha amino acid group Chemical group 0.000 claims description 135
- 238000012360 testing method Methods 0.000 claims description 66
- 239000003814 drug Substances 0.000 claims description 63
- 229940079593 drug Drugs 0.000 claims description 61
- 101000629313 Severe acute respiratory syndrome coronavirus Spike glycoprotein Proteins 0.000 claims description 53
- 208000024891 symptom Diseases 0.000 claims description 53
- 241000282414 Homo sapiens Species 0.000 claims description 51
- 101000629318 Severe acute respiratory syndrome coronavirus 2 Spike glycoprotein Proteins 0.000 claims description 40
- 208000025721 COVID-19 Diseases 0.000 claims description 33
- 208000015181 infectious disease Diseases 0.000 claims description 32
- 230000010530 Virus Neutralization Effects 0.000 claims description 30
- 101710198474 Spike protein Proteins 0.000 claims description 24
- 229940096437 Protein S Drugs 0.000 claims description 23
- 229960005486 vaccine Drugs 0.000 claims description 23
- 230000009467 reduction Effects 0.000 claims description 21
- 208000001528 Coronaviridae Infections Diseases 0.000 claims description 19
- 241000700605 Viruses Species 0.000 claims description 19
- 229940022962 COVID-19 vaccine Drugs 0.000 claims description 17
- 206010061598 Immunodeficiency Diseases 0.000 claims description 15
- 230000001965 increasing effect Effects 0.000 claims description 14
- 230000003612 virological effect Effects 0.000 claims description 14
- 230000001684 chronic effect Effects 0.000 claims description 13
- 239000007787 solid Substances 0.000 claims description 13
- 210000004369 blood Anatomy 0.000 claims description 12
- 239000008280 blood Substances 0.000 claims description 12
- 206010012601 diabetes mellitus Diseases 0.000 claims description 12
- 208000008589 Obesity Diseases 0.000 claims description 11
- 206010035664 Pneumonia Diseases 0.000 claims description 11
- 208000006673 asthma Diseases 0.000 claims description 11
- 235000020824 obesity Nutrition 0.000 claims description 11
- 210000000056 organ Anatomy 0.000 claims description 11
- 206010061818 Disease progression Diseases 0.000 claims description 10
- 206010020772 Hypertension Diseases 0.000 claims description 10
- 230000003247 decreasing effect Effects 0.000 claims description 10
- 230000005750 disease progression Effects 0.000 claims description 10
- 239000003018 immunosuppressive agent Substances 0.000 claims description 10
- 230000001404 mediated effect Effects 0.000 claims description 10
- 230000035935 pregnancy Effects 0.000 claims description 10
- 230000003442 weekly effect Effects 0.000 claims description 10
- 208000020832 chronic kidney disease Diseases 0.000 claims description 9
- 238000009826 distribution Methods 0.000 claims description 9
- 230000028993 immune response Effects 0.000 claims description 9
- 229960003444 immunosuppressant agent Drugs 0.000 claims description 9
- 230000001861 immunosuppressant effect Effects 0.000 claims description 9
- 230000001939 inductive effect Effects 0.000 claims description 9
- 241000894006 Bacteria Species 0.000 claims description 8
- 201000010374 Down Syndrome Diseases 0.000 claims description 8
- 208000029462 Immunodeficiency disease Diseases 0.000 claims description 8
- 102220599672 Spindlin-1_D614G_mutation Human genes 0.000 claims description 8
- 206010043395 Thalassaemia sickle cell Diseases 0.000 claims description 8
- 208000007536 Thrombosis Diseases 0.000 claims description 8
- 230000001363 autoimmune Effects 0.000 claims description 8
- 230000000747 cardiac effect Effects 0.000 claims description 8
- 206010014599 encephalitis Diseases 0.000 claims description 8
- 208000019622 heart disease Diseases 0.000 claims description 8
- 230000007813 immunodeficiency Effects 0.000 claims description 8
- 201000004193 respiratory failure Diseases 0.000 claims description 8
- 208000007056 sickle cell anemia Diseases 0.000 claims description 8
- 210000000130 stem cell Anatomy 0.000 claims description 8
- 208000011117 substance-related disease Diseases 0.000 claims description 8
- 238000002560 therapeutic procedure Methods 0.000 claims description 8
- 208000026106 cerebrovascular disease Diseases 0.000 claims description 7
- 230000034994 death Effects 0.000 claims description 7
- 231100000517 death Toxicity 0.000 claims description 7
- 208000023516 stroke disease Diseases 0.000 claims description 7
- 206010006451 bronchitis Diseases 0.000 claims description 6
- 206010052015 cytokine release syndrome Diseases 0.000 claims description 6
- 230000002496 gastric effect Effects 0.000 claims description 6
- 230000002757 inflammatory effect Effects 0.000 claims description 6
- 238000005399 mechanical ventilation Methods 0.000 claims description 6
- 206010019280 Heart failures Diseases 0.000 claims description 5
- 208000019693 Lung disease Diseases 0.000 claims description 5
- 230000001010 compromised effect Effects 0.000 claims description 5
- 239000012530 fluid Substances 0.000 claims description 5
- 230000000241 respiratory effect Effects 0.000 claims description 5
- 208000009304 Acute Kidney Injury Diseases 0.000 claims description 4
- 206010001052 Acute respiratory distress syndrome Diseases 0.000 claims description 4
- 206010002199 Anaphylactic shock Diseases 0.000 claims description 4
- 201000002909 Aspergillosis Diseases 0.000 claims description 4
- 208000036641 Aspergillus infections Diseases 0.000 claims description 4
- 208000014644 Brain disease Diseases 0.000 claims description 4
- 208000011380 COVID-19–associated multisystem inflammatory syndrome in children Diseases 0.000 claims description 4
- 208000020446 Cardiac disease Diseases 0.000 claims description 4
- 206010008111 Cerebral haemorrhage Diseases 0.000 claims description 4
- 206010010356 Congenital anomaly Diseases 0.000 claims description 4
- 206010010904 Convulsion Diseases 0.000 claims description 4
- 206010014522 Embolism venous Diseases 0.000 claims description 4
- 208000032096 Encephalitic infection Diseases 0.000 claims description 4
- 208000032274 Encephalopathy Diseases 0.000 claims description 4
- 208000010496 Heart Arrest Diseases 0.000 claims description 4
- 208000028622 Immune thrombocytopenia Diseases 0.000 claims description 4
- 208000032382 Ischaemic stroke Diseases 0.000 claims description 4
- 208000034800 Leukoencephalopathies Diseases 0.000 claims description 4
- 206010067125 Liver injury Diseases 0.000 claims description 4
- 208000009525 Myocarditis Diseases 0.000 claims description 4
- 208000029726 Neurodevelopmental disease Diseases 0.000 claims description 4
- 206010053159 Organ failure Diseases 0.000 claims description 4
- 206010033627 Pancreatic injury Diseases 0.000 claims description 4
- 208000028017 Psychotic disease Diseases 0.000 claims description 4
- 208000033626 Renal failure acute Diseases 0.000 claims description 4
- 208000013616 Respiratory Distress Syndrome Diseases 0.000 claims description 4
- 208000004756 Respiratory Insufficiency Diseases 0.000 claims description 4
- 206010040070 Septic Shock Diseases 0.000 claims description 4
- 208000006011 Stroke Diseases 0.000 claims description 4
- 208000031981 Thrombocytopenic Idiopathic Purpura Diseases 0.000 claims description 4
- 206010043784 Thyroiditis subacute Diseases 0.000 claims description 4
- 208000030886 Traumatic Brain injury Diseases 0.000 claims description 4
- 208000002552 acute disseminated encephalomyelitis Diseases 0.000 claims description 4
- 201000011040 acute kidney failure Diseases 0.000 claims description 4
- 231100000439 acute liver injury Toxicity 0.000 claims description 4
- 206010000891 acute myocardial infarction Diseases 0.000 claims description 4
- 206010001053 acute respiratory failure Diseases 0.000 claims description 4
- 201000000028 adult respiratory distress syndrome Diseases 0.000 claims description 4
- 208000003455 anaphylaxis Diseases 0.000 claims description 4
- 230000009517 anoxic brain damage Effects 0.000 claims description 4
- 206010003119 arrhythmia Diseases 0.000 claims description 4
- 201000003710 autoimmune thrombocytopenic purpura Diseases 0.000 claims description 4
- 208000009190 disseminated intravascular coagulation Diseases 0.000 claims description 4
- 230000002526 effect on cardiovascular system Effects 0.000 claims description 4
- 208000020658 intracerebral hemorrhage Diseases 0.000 claims description 4
- 208000037891 myocardial injury Diseases 0.000 claims description 4
- 230000002956 necrotizing effect Effects 0.000 claims description 4
- 208000022076 postinfectious encephalitis Diseases 0.000 claims description 4
- 230000002441 reversible effect Effects 0.000 claims description 4
- 230000036303 septic shock Effects 0.000 claims description 4
- 230000035939 shock Effects 0.000 claims description 4
- 208000005809 status epilepticus Diseases 0.000 claims description 4
- 201000007497 subacute thyroiditis Diseases 0.000 claims description 4
- 208000011580 syndromic disease Diseases 0.000 claims description 4
- 201000003067 thrombocytopenia due to platelet alloimmunization Diseases 0.000 claims description 4
- 230000009529 traumatic brain injury Effects 0.000 claims description 4
- 208000004043 venous thromboembolism Diseases 0.000 claims description 4
- 208000014085 Chronic respiratory disease Diseases 0.000 claims description 2
- 102100031673 Corneodesmosin Human genes 0.000 abstract description 53
- 101710139375 Corneodesmosin Proteins 0.000 abstract description 53
- 238000011282 treatment Methods 0.000 abstract description 16
- 230000009286 beneficial effect Effects 0.000 abstract description 6
- 238000003745 diagnosis Methods 0.000 abstract 1
- 208000037765 diseases and disorders Diseases 0.000 abstract 1
- 230000005764 inhibitory process Effects 0.000 abstract 1
- 108010047041 Complementarity Determining Regions Proteins 0.000 description 228
- 210000004027 cell Anatomy 0.000 description 201
- 102000008394 Immunoglobulin Fragments Human genes 0.000 description 113
- 108010021625 Immunoglobulin Fragments Proteins 0.000 description 113
- 108090000765 processed proteins & peptides Proteins 0.000 description 71
- 239000000523 sample Substances 0.000 description 71
- 150000007523 nucleic acids Chemical class 0.000 description 66
- 235000018102 proteins Nutrition 0.000 description 63
- 102000004196 processed proteins & peptides Human genes 0.000 description 55
- 108091028043 Nucleic acid sequence Proteins 0.000 description 53
- 235000001014 amino acid Nutrition 0.000 description 53
- 229920001184 polypeptide Polymers 0.000 description 51
- 150000001413 amino acids Chemical class 0.000 description 39
- 102000005962 receptors Human genes 0.000 description 39
- 108020003175 receptors Proteins 0.000 description 39
- 239000013598 vector Substances 0.000 description 38
- 241000127282 Middle East respiratory syndrome-related coronavirus Species 0.000 description 36
- 201000003176 Severe Acute Respiratory Syndrome Diseases 0.000 description 36
- 238000003556 assay Methods 0.000 description 32
- 238000006467 substitution reaction Methods 0.000 description 31
- 230000014509 gene expression Effects 0.000 description 30
- 108020004414 DNA Proteins 0.000 description 26
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 25
- 238000007918 intramuscular administration Methods 0.000 description 25
- 102100035765 Angiotensin-converting enzyme 2 Human genes 0.000 description 22
- 108090000975 Angiotensin-converting enzyme 2 Proteins 0.000 description 22
- 210000004962 mammalian cell Anatomy 0.000 description 21
- 240000004808 Saccharomyces cerevisiae Species 0.000 description 19
- -1 ciclesonide Chemical class 0.000 description 19
- 239000000203 mixture Substances 0.000 description 19
- 241000711467 Human coronavirus 229E Species 0.000 description 18
- 201000010099 disease Diseases 0.000 description 18
- 230000002829 reductive effect Effects 0.000 description 18
- 241000482741 Human coronavirus NL63 Species 0.000 description 17
- 108091026890 Coding region Proteins 0.000 description 16
- 102100025012 Dipeptidyl peptidase 4 Human genes 0.000 description 16
- 206010061218 Inflammation Diseases 0.000 description 16
- 239000013604 expression vector Substances 0.000 description 16
- 230000004054 inflammatory process Effects 0.000 description 16
- 239000003550 marker Substances 0.000 description 16
- 102000040430 polynucleotide Human genes 0.000 description 16
- 108091033319 polynucleotide Proteins 0.000 description 16
- 239000002157 polynucleotide Substances 0.000 description 16
- 238000013518 transcription Methods 0.000 description 16
- 230000035897 transcription Effects 0.000 description 16
- 102100040843 C-type lectin domain family 4 member M Human genes 0.000 description 15
- 101000749311 Homo sapiens C-type lectin domain family 4 member M Proteins 0.000 description 15
- 108010076504 Protein Sorting Signals Proteins 0.000 description 15
- 101000908391 Homo sapiens Dipeptidyl peptidase 4 Proteins 0.000 description 14
- 241001465754 Metazoa Species 0.000 description 14
- 125000000539 amino acid group Chemical group 0.000 description 14
- 230000006870 function Effects 0.000 description 14
- 238000004519 manufacturing process Methods 0.000 description 14
- 241001428935 Human coronavirus OC43 Species 0.000 description 13
- 239000012636 effector Substances 0.000 description 13
- 230000000694 effects Effects 0.000 description 13
- 102000039446 nucleic acids Human genes 0.000 description 13
- 108020004707 nucleic acids Proteins 0.000 description 13
- 229920001223 polyethylene glycol Polymers 0.000 description 13
- 108010087819 Fc receptors Proteins 0.000 description 12
- 102000009109 Fc receptors Human genes 0.000 description 12
- 102100026120 IgG receptor FcRn large subunit p51 Human genes 0.000 description 12
- 108060003951 Immunoglobulin Proteins 0.000 description 12
- 102000018358 immunoglobulin Human genes 0.000 description 12
- 238000001990 intravenous administration Methods 0.000 description 12
- 101710177940 IgG receptor FcRn large subunit p51 Proteins 0.000 description 11
- 239000002202 Polyethylene glycol Substances 0.000 description 11
- 210000004072 lung Anatomy 0.000 description 11
- 230000006798 recombination Effects 0.000 description 11
- 238000005215 recombination Methods 0.000 description 11
- 241000829100 Macaca mulatta polyomavirus 1 Species 0.000 description 10
- 230000004071 biological effect Effects 0.000 description 10
- 238000011161 development Methods 0.000 description 10
- 239000003623 enhancer Substances 0.000 description 10
- 230000035772 mutation Effects 0.000 description 10
- 230000010076 replication Effects 0.000 description 10
- 108010019670 Chimeric Antigen Receptors Proteins 0.000 description 9
- 102100024952 Protein CBFA2T1 Human genes 0.000 description 9
- 238000012867 alanine scanning Methods 0.000 description 9
- 229940049595 antibody-drug conjugate Drugs 0.000 description 9
- 238000010367 cloning Methods 0.000 description 9
- 238000001727 in vivo Methods 0.000 description 9
- 230000003993 interaction Effects 0.000 description 9
- 210000003292 kidney cell Anatomy 0.000 description 9
- 238000013507 mapping Methods 0.000 description 9
- 230000037452 priming Effects 0.000 description 9
- 230000001225 therapeutic effect Effects 0.000 description 9
- 206010011224 Cough Diseases 0.000 description 8
- 108010054477 Immunoglobulin Fab Fragments Proteins 0.000 description 8
- 102000001706 Immunoglobulin Fab Fragments Human genes 0.000 description 8
- 239000003443 antiviral agent Substances 0.000 description 8
- 230000000875 corresponding effect Effects 0.000 description 8
- 230000036541 health Effects 0.000 description 8
- 239000003112 inhibitor Substances 0.000 description 8
- 230000002401 inhibitory effect Effects 0.000 description 8
- 238000002372 labelling Methods 0.000 description 8
- 239000002773 nucleotide Substances 0.000 description 8
- 125000003729 nucleotide group Chemical group 0.000 description 8
- 210000000689 upper leg Anatomy 0.000 description 8
- 238000002255 vaccination Methods 0.000 description 8
- 210000002845 virion Anatomy 0.000 description 8
- 102100024746 Dihydrofolate reductase Human genes 0.000 description 7
- 102000004190 Enzymes Human genes 0.000 description 7
- 108090000790 Enzymes Proteins 0.000 description 7
- 241000238631 Hexapoda Species 0.000 description 7
- 101000638154 Homo sapiens Transmembrane protease serine 2 Proteins 0.000 description 7
- 108090001074 Nucleocapsid Proteins Proteins 0.000 description 7
- 241001112090 Pseudovirus Species 0.000 description 7
- 206010037660 Pyrexia Diseases 0.000 description 7
- 239000002260 anti-inflammatory agent Substances 0.000 description 7
- 230000010056 antibody-dependent cellular cytotoxicity Effects 0.000 description 7
- 230000000890 antigenic effect Effects 0.000 description 7
- 238000012258 culturing Methods 0.000 description 7
- 108020001096 dihydrofolate reductase Proteins 0.000 description 7
- 229940090124 dipeptidyl peptidase 4 (dpp-4) inhibitors for blood glucose lowering Drugs 0.000 description 7
- 208000035475 disorder Diseases 0.000 description 7
- 229940088598 enzyme Drugs 0.000 description 7
- 239000013612 plasmid Substances 0.000 description 7
- RWWYLEGWBNMMLJ-MEUHYHILSA-N remdesivir Drugs C([C@@H]1[C@H]([C@@H](O)[C@@](C#N)(O1)C=1N2N=CN=C(N)C2=CC=1)O)OP(=O)(N[C@@H](C)C(=O)OCC(CC)CC)OC1=CC=CC=C1 RWWYLEGWBNMMLJ-MEUHYHILSA-N 0.000 description 7
- RWWYLEGWBNMMLJ-YSOARWBDSA-N remdesivir Chemical compound NC1=NC=NN2C1=CC=C2[C@]1([C@@H]([C@@H]([C@H](O1)CO[P@](=O)(OC1=CC=CC=C1)N[C@H](C(=O)OCC(CC)CC)C)O)O)C#N RWWYLEGWBNMMLJ-YSOARWBDSA-N 0.000 description 7
- 210000005253 yeast cell Anatomy 0.000 description 7
- 102000036364 Cullin Ring E3 Ligases Human genes 0.000 description 6
- 239000004365 Protease Substances 0.000 description 6
- 238000004458 analytical method Methods 0.000 description 6
- 239000003795 chemical substances by application Substances 0.000 description 6
- 230000029087 digestion Effects 0.000 description 6
- ZCGNOVWYSGBHAU-UHFFFAOYSA-N favipiravir Chemical compound NC(=O)C1=NC(F)=CNC1=O ZCGNOVWYSGBHAU-UHFFFAOYSA-N 0.000 description 6
- 230000012010 growth Effects 0.000 description 6
- 239000001963 growth medium Substances 0.000 description 6
- 230000001900 immune effect Effects 0.000 description 6
- 229940072221 immunoglobulins Drugs 0.000 description 6
- 238000000338 in vitro Methods 0.000 description 6
- 239000000463 material Substances 0.000 description 6
- 239000002609 medium Substances 0.000 description 6
- 235000015097 nutrients Nutrition 0.000 description 6
- 230000004044 response Effects 0.000 description 6
- AZSNMRSAGSSBNP-UHFFFAOYSA-N 22,23-dihydroavermectin B1a Natural products C1CC(C)C(C(C)CC)OC21OC(CC=C(C)C(OC1OC(C)C(OC3OC(C)C(O)C(OC)C3)C(OC)C1)C(C)C=CC=C1C3(C(C(=O)O4)C=C(C)C(O)C3OC1)O)CC4C2 AZSNMRSAGSSBNP-UHFFFAOYSA-N 0.000 description 5
- SPBDXSGPUHCETR-JFUDTMANSA-N 8883yp2r6d Chemical compound O1[C@@H](C)[C@H](O)[C@@H](OC)C[C@@H]1O[C@@H]1[C@@H](OC)C[C@H](O[C@@H]2C(=C/C[C@@H]3C[C@@H](C[C@@]4(O[C@@H]([C@@H](C)CC4)C(C)C)O3)OC(=O)[C@@H]3C=C(C)[C@@H](O)[C@H]4OC\C([C@@]34O)=C/C=C/[C@@H]2C)/C)O[C@H]1C.C1C[C@H](C)[C@@H]([C@@H](C)CC)O[C@@]21O[C@H](C\C=C(C)\[C@@H](O[C@@H]1O[C@@H](C)[C@H](O[C@@H]3O[C@@H](C)[C@H](O)[C@@H](OC)C3)[C@@H](OC)C1)[C@@H](C)\C=C\C=C/1[C@]3([C@H](C(=O)O4)C=C(C)[C@@H](O)[C@H]3OC\1)O)C[C@H]4C2 SPBDXSGPUHCETR-JFUDTMANSA-N 0.000 description 5
- QAGYKUNXZHXKMR-UHFFFAOYSA-N CPD000469186 Natural products CC1=C(O)C=CC=C1C(=O)NC(C(O)CN1C(CC2CCCCC2C1)C(=O)NC(C)(C)C)CSC1=CC=CC=C1 QAGYKUNXZHXKMR-UHFFFAOYSA-N 0.000 description 5
- 241000494545 Cordyline virus 2 Species 0.000 description 5
- 208000000059 Dyspnea Diseases 0.000 description 5
- 206010013975 Dyspnoeas Diseases 0.000 description 5
- 241000282412 Homo Species 0.000 description 5
- KJHKTHWMRKYKJE-SUGCFTRWSA-N Kaletra Chemical compound N1([C@@H](C(C)C)C(=O)N[C@H](C[C@H](O)[C@H](CC=2C=CC=CC=2)NC(=O)COC=2C(=CC=CC=2C)C)CC=2C=CC=CC=2)CCCNC1=O KJHKTHWMRKYKJE-SUGCFTRWSA-N 0.000 description 5
- QNAYBMKLOCPYGJ-REOHCLBHSA-N L-alanine Chemical compound C[C@H](N)C(O)=O QNAYBMKLOCPYGJ-REOHCLBHSA-N 0.000 description 5
- FBOZXECLQNJBKD-ZDUSSCGKSA-N L-methotrexate Chemical compound C=1N=C2N=C(N)N=C(N)C2=NC=1CN(C)C1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 FBOZXECLQNJBKD-ZDUSSCGKSA-N 0.000 description 5
- 229940026207 Moderna COVID-19 vaccine Drugs 0.000 description 5
- NCDNCNXCDXHOMX-UHFFFAOYSA-N Ritonavir Natural products C=1C=CC=CC=1CC(NC(=O)OCC=1SC=NC=1)C(O)CC(CC=1C=CC=CC=1)NC(=O)C(C(C)C)NC(=O)N(C)CC1=CSC(C(C)C)=N1 NCDNCNXCDXHOMX-UHFFFAOYSA-N 0.000 description 5
- 102100031989 Transmembrane protease serine 2 Human genes 0.000 description 5
- 235000004279 alanine Nutrition 0.000 description 5
- 229940124599 anti-inflammatory drug Drugs 0.000 description 5
- 239000000611 antibody drug conjugate Substances 0.000 description 5
- 230000001580 bacterial effect Effects 0.000 description 5
- 230000008859 change Effects 0.000 description 5
- 210000004978 chinese hamster ovary cell Anatomy 0.000 description 5
- 230000000295 complement effect Effects 0.000 description 5
- 239000002299 complementary DNA Substances 0.000 description 5
- 229960005107 darunavir Drugs 0.000 description 5
- CJBJHOAVZSMMDJ-HEXNFIEUSA-N darunavir Chemical compound C([C@@H]([C@H](O)CN(CC(C)C)S(=O)(=O)C=1C=CC(N)=CC=1)NC(=O)O[C@@H]1[C@@H]2CCO[C@@H]2OC1)C1=CC=CC=C1 CJBJHOAVZSMMDJ-HEXNFIEUSA-N 0.000 description 5
- 229950008454 favipiravir Drugs 0.000 description 5
- 230000002538 fungal effect Effects 0.000 description 5
- 230000004927 fusion Effects 0.000 description 5
- 238000003364 immunohistochemistry Methods 0.000 description 5
- 229960002418 ivermectin Drugs 0.000 description 5
- 229960004525 lopinavir Drugs 0.000 description 5
- 229960000884 nelfinavir Drugs 0.000 description 5
- QAGYKUNXZHXKMR-HKWSIXNMSA-N nelfinavir Chemical compound CC1=C(O)C=CC=C1C(=O)N[C@H]([C@H](O)CN1[C@@H](C[C@@H]2CCCC[C@@H]2C1)C(=O)NC(C)(C)C)CSC1=CC=CC=C1 QAGYKUNXZHXKMR-HKWSIXNMSA-N 0.000 description 5
- 230000002265 prevention Effects 0.000 description 5
- 230000000069 prophylactic effect Effects 0.000 description 5
- 229960000311 ritonavir Drugs 0.000 description 5
- NCDNCNXCDXHOMX-XGKFQTDJSA-N ritonavir Chemical compound N([C@@H](C(C)C)C(=O)N[C@H](C[C@H](O)[C@H](CC=1C=CC=CC=1)NC(=O)OCC=1SC=NC=1)CC=1C=CC=CC=1)C(=O)N(C)CC1=CSC(C(C)C)=N1 NCDNCNXCDXHOMX-XGKFQTDJSA-N 0.000 description 5
- 229960001852 saquinavir Drugs 0.000 description 5
- QWAXKHKRTORLEM-UGJKXSETSA-N saquinavir Chemical compound C([C@@H]([C@H](O)CN1C[C@H]2CCCC[C@H]2C[C@H]1C(=O)NC(C)(C)C)NC(=O)[C@H](CC(N)=O)NC(=O)C=1N=C2C=CC=CC2=CC=1)C1=CC=CC=C1 QWAXKHKRTORLEM-UGJKXSETSA-N 0.000 description 5
- 208000013220 shortness of breath Diseases 0.000 description 5
- 241000894007 species Species 0.000 description 5
- 239000000126 substance Substances 0.000 description 5
- 230000004083 survival effect Effects 0.000 description 5
- 230000005727 virus proliferation Effects 0.000 description 5
- YBJHBAHKTGYVGT-ZKWXMUAHSA-N (+)-Biotin Chemical compound N1C(=O)N[C@@H]2[C@H](CCCCC(=O)O)SC[C@@H]21 YBJHBAHKTGYVGT-ZKWXMUAHSA-N 0.000 description 4
- WHTVZRBIWZFKQO-AWEZNQCLSA-N (S)-chloroquine Chemical compound ClC1=CC=C2C(N[C@@H](C)CCCN(CC)CC)=CC=NC2=C1 WHTVZRBIWZFKQO-AWEZNQCLSA-N 0.000 description 4
- 239000005541 ACE inhibitor Substances 0.000 description 4
- LUKZNWIVRBCLON-GXOBDPJESA-N Ciclesonide Chemical compound C1([C@H]2O[C@@]3([C@H](O2)C[C@@H]2[C@@]3(C[C@H](O)[C@@H]3[C@@]4(C)C=CC(=O)C=C4CC[C@H]32)C)C(=O)COC(=O)C(C)C)CCCCC1 LUKZNWIVRBCLON-GXOBDPJESA-N 0.000 description 4
- 241000699800 Cricetinae Species 0.000 description 4
- 241000699802 Cricetulus griseus Species 0.000 description 4
- 102000004127 Cytokines Human genes 0.000 description 4
- 108090000695 Cytokines Proteins 0.000 description 4
- 108010008165 Etanercept Proteins 0.000 description 4
- 108700028146 Genetic Enhancer Elements Proteins 0.000 description 4
- 244000309467 Human Coronavirus Species 0.000 description 4
- 108091006905 Human Serum Albumin Proteins 0.000 description 4
- 102000008100 Human Serum Albumin Human genes 0.000 description 4
- WHUUTDBJXJRKMK-VKHMYHEASA-N L-glutamic acid Chemical compound OC(=O)[C@@H](N)CCC(O)=O WHUUTDBJXJRKMK-VKHMYHEASA-N 0.000 description 4
- KDXKERNSBIXSRK-UHFFFAOYSA-N Lysine Natural products NCCCCC(N)C(O)=O KDXKERNSBIXSRK-UHFFFAOYSA-N 0.000 description 4
- 239000004472 Lysine Substances 0.000 description 4
- 108090000157 Metallothionein Proteins 0.000 description 4
- 208000025370 Middle East respiratory syndrome Diseases 0.000 description 4
- 241000699666 Mus <mouse, genus> Species 0.000 description 4
- 229930193140 Neomycin Natural products 0.000 description 4
- 229940025109 Oxford–AstraZeneca COVID-19 vaccine Drugs 0.000 description 4
- 102000035195 Peptidases Human genes 0.000 description 4
- 108091005804 Peptidases Proteins 0.000 description 4
- 229940122055 Serine protease inhibitor Drugs 0.000 description 4
- 101710102218 Serine protease inhibitor Proteins 0.000 description 4
- 101710172711 Structural protein Proteins 0.000 description 4
- 108090000631 Trypsin Proteins 0.000 description 4
- 102000004142 Trypsin Human genes 0.000 description 4
- 108060008683 Tumor Necrosis Factor Receptor Proteins 0.000 description 4
- 230000004913 activation Effects 0.000 description 4
- 229960002964 adalimumab Drugs 0.000 description 4
- 230000009824 affinity maturation Effects 0.000 description 4
- 229940044094 angiotensin-converting-enzyme inhibitor Drugs 0.000 description 4
- 239000003242 anti bacterial agent Substances 0.000 description 4
- 239000000739 antihistaminic agent Substances 0.000 description 4
- 229960003159 atovaquone Drugs 0.000 description 4
- KUCQYCKVKVOKAY-CTYIDZIISA-N atovaquone Chemical compound C1([C@H]2CC[C@@H](CC2)C2=C(C(C3=CC=CC=C3C2=O)=O)O)=CC=C(Cl)C=C1 KUCQYCKVKVOKAY-CTYIDZIISA-N 0.000 description 4
- 210000003719 b-lymphocyte Anatomy 0.000 description 4
- 230000003115 biocidal effect Effects 0.000 description 4
- 239000006143 cell culture medium Substances 0.000 description 4
- 238000012512 characterization method Methods 0.000 description 4
- 229960003677 chloroquine Drugs 0.000 description 4
- WHTVZRBIWZFKQO-UHFFFAOYSA-N chloroquine Natural products ClC1=CC=C2C(NC(C)CCCN(CC)CC)=CC=NC2=C1 WHTVZRBIWZFKQO-UHFFFAOYSA-N 0.000 description 4
- 229960003728 ciclesonide Drugs 0.000 description 4
- 239000003246 corticosteroid Substances 0.000 description 4
- 229960001334 corticosteroids Drugs 0.000 description 4
- 238000012217 deletion Methods 0.000 description 4
- 230000037430 deletion Effects 0.000 description 4
- 229960000403 etanercept Drugs 0.000 description 4
- 238000002474 experimental method Methods 0.000 description 4
- XXSMGPRMXLTPCZ-UHFFFAOYSA-N hydroxychloroquine Chemical compound ClC1=CC=C2C(NC(C)CCCN(CCO)CC)=CC=NC2=C1 XXSMGPRMXLTPCZ-UHFFFAOYSA-N 0.000 description 4
- 229960004171 hydroxychloroquine Drugs 0.000 description 4
- NOESYZHRGYRDHS-UHFFFAOYSA-N insulin Chemical compound N1C(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(NC(=O)CN)C(C)CC)CSSCC(C(NC(CO)C(=O)NC(CC(C)C)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CCC(N)=O)C(=O)NC(CC(C)C)C(=O)NC(CCC(O)=O)C(=O)NC(CC(N)=O)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CSSCC(NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2C=CC(O)=CC=2)NC(=O)C(CC(C)C)NC(=O)C(C)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2NC=NC=2)NC(=O)C(CO)NC(=O)CNC2=O)C(=O)NCC(=O)NC(CCC(O)=O)C(=O)NC(CCCNC(N)=N)C(=O)NCC(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC(O)=CC=3)C(=O)NC(C(C)O)C(=O)N3C(CCC3)C(=O)NC(CCCCN)C(=O)NC(C)C(O)=O)C(=O)NC(CC(N)=O)C(O)=O)=O)NC(=O)C(C(C)CC)NC(=O)C(CO)NC(=O)C(C(C)O)NC(=O)C1CSSCC2NC(=O)C(CC(C)C)NC(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CC(N)=O)NC(=O)C(NC(=O)C(N)CC=1C=CC=CC=1)C(C)C)CC1=CN=CN1 NOESYZHRGYRDHS-UHFFFAOYSA-N 0.000 description 4
- 210000005229 liver cell Anatomy 0.000 description 4
- 235000018977 lysine Nutrition 0.000 description 4
- 229960000485 methotrexate Drugs 0.000 description 4
- 230000004048 modification Effects 0.000 description 4
- 238000012986 modification Methods 0.000 description 4
- 238000002703 mutagenesis Methods 0.000 description 4
- 231100000350 mutagenesis Toxicity 0.000 description 4
- 229960004927 neomycin Drugs 0.000 description 4
- 239000008194 pharmaceutical composition Substances 0.000 description 4
- 230000008569 process Effects 0.000 description 4
- 239000012857 radioactive material Substances 0.000 description 4
- 230000001105 regulatory effect Effects 0.000 description 4
- 230000028327 secretion Effects 0.000 description 4
- 239000003001 serine protease inhibitor Substances 0.000 description 4
- 238000002741 site-directed mutagenesis Methods 0.000 description 4
- 238000001228 spectrum Methods 0.000 description 4
- 150000003431 steroids Chemical class 0.000 description 4
- 239000013589 supplement Substances 0.000 description 4
- 210000001519 tissue Anatomy 0.000 description 4
- 239000003053 toxin Substances 0.000 description 4
- 231100000765 toxin Toxicity 0.000 description 4
- 108700012359 toxins Proteins 0.000 description 4
- 230000005030 transcription termination Effects 0.000 description 4
- 230000014616 translation Effects 0.000 description 4
- 239000012588 trypsin Substances 0.000 description 4
- 229960002109 tuberculosis vaccine Drugs 0.000 description 4
- 229940046728 tumor necrosis factor alpha inhibitor Drugs 0.000 description 4
- 239000002451 tumor necrosis factor inhibitor Substances 0.000 description 4
- 102000003298 tumor necrosis factor receptor Human genes 0.000 description 4
- 241000701161 unidentified adenovirus Species 0.000 description 4
- MFRNYXJJRJQHNW-DEMKXPNLSA-N (2s)-2-[[(2r,3r)-3-methoxy-3-[(2s)-1-[(3r,4s,5s)-3-methoxy-5-methyl-4-[methyl-[(2s)-3-methyl-2-[[(2s)-3-methyl-2-(methylamino)butanoyl]amino]butanoyl]amino]heptanoyl]pyrrolidin-2-yl]-2-methylpropanoyl]amino]-3-phenylpropanoic acid Chemical compound CN[C@@H](C(C)C)C(=O)N[C@@H](C(C)C)C(=O)N(C)[C@@H]([C@@H](C)CC)[C@H](OC)CC(=O)N1CCC[C@H]1[C@H](OC)[C@@H](C)C(=O)N[C@H](C(O)=O)CC1=CC=CC=C1 MFRNYXJJRJQHNW-DEMKXPNLSA-N 0.000 description 3
- 229940125678 AZD7442 Drugs 0.000 description 3
- 241000004176 Alphacoronavirus Species 0.000 description 3
- 241000271566 Aves Species 0.000 description 3
- 241000008921 Avian coronavirus Species 0.000 description 3
- 241000282693 Cercopithecidae Species 0.000 description 3
- 108700022167 ChAdOx1 nCoV-19 Proteins 0.000 description 3
- 206010010741 Conjunctivitis Diseases 0.000 description 3
- 201000003883 Cystic fibrosis Diseases 0.000 description 3
- 241000701022 Cytomegalovirus Species 0.000 description 3
- 238000002965 ELISA Methods 0.000 description 3
- LYCAIKOWRPUZTN-UHFFFAOYSA-N Ethylene glycol Chemical compound OCCO LYCAIKOWRPUZTN-UHFFFAOYSA-N 0.000 description 3
- WHUUTDBJXJRKMK-UHFFFAOYSA-N Glutamic acid Natural products OC(=O)C(N)CCC(O)=O WHUUTDBJXJRKMK-UHFFFAOYSA-N 0.000 description 3
- 208000031886 HIV Infections Diseases 0.000 description 3
- 208000037357 HIV infectious disease Diseases 0.000 description 3
- 229940124790 IL-6 inhibitor Drugs 0.000 description 3
- 108010038501 Interleukin-6 Receptors Proteins 0.000 description 3
- 102000010781 Interleukin-6 Receptors Human genes 0.000 description 3
- CKLJMWTZIZZHCS-REOHCLBHSA-N L-aspartic acid Chemical compound OC(=O)[C@@H](N)CC(O)=O CKLJMWTZIZZHCS-REOHCLBHSA-N 0.000 description 3
- HNDVDQJCIGZPNO-YFKPBYRVSA-N L-histidine Chemical compound OC(=O)[C@@H](N)CC1=CN=CN1 HNDVDQJCIGZPNO-YFKPBYRVSA-N 0.000 description 3
- KDXKERNSBIXSRK-YFKPBYRVSA-N L-lysine Chemical compound NCCCC[C@H](N)C(O)=O KDXKERNSBIXSRK-YFKPBYRVSA-N 0.000 description 3
- 241000282560 Macaca mulatta Species 0.000 description 3
- UWWDHYUMIORJTA-HSQYWUDLSA-N Moexipril Chemical compound C([C@@H](C(=O)OCC)N[C@@H](C)C(=O)N1[C@@H](CC2=CC(OC)=C(OC)C=C2C1)C(O)=O)CC1=CC=CC=C1 UWWDHYUMIORJTA-HSQYWUDLSA-N 0.000 description 3
- 238000005481 NMR spectroscopy Methods 0.000 description 3
- 108700026244 Open Reading Frames Proteins 0.000 description 3
- 229940026233 Pfizer-BioNTech COVID-19 vaccine Drugs 0.000 description 3
- 206010057249 Phagocytosis Diseases 0.000 description 3
- 108010008281 Recombinant Fusion Proteins Proteins 0.000 description 3
- 102000007056 Recombinant Fusion Proteins Human genes 0.000 description 3
- 108010003723 Single-Domain Antibodies Proteins 0.000 description 3
- 241000251539 Vertebrata <Metazoa> Species 0.000 description 3
- 230000002378 acidificating effect Effects 0.000 description 3
- 238000007792 addition Methods 0.000 description 3
- 230000003321 amplification Effects 0.000 description 3
- 239000005557 antagonist Substances 0.000 description 3
- 230000000844 anti-bacterial effect Effects 0.000 description 3
- 229940088710 antibiotic agent Drugs 0.000 description 3
- 210000000628 antibody-producing cell Anatomy 0.000 description 3
- 229940124623 antihistamine drug Drugs 0.000 description 3
- 239000003096 antiparasitic agent Substances 0.000 description 3
- 235000003704 aspartic acid Nutrition 0.000 description 3
- 230000008901 benefit Effects 0.000 description 3
- YWGDOWXRIALTES-NRFANRHFSA-N bepotastine Chemical compound C1CN(CCCC(=O)O)CCC1O[C@H](C=1N=CC=CC=1)C1=CC=C(Cl)C=C1 YWGDOWXRIALTES-NRFANRHFSA-N 0.000 description 3
- 229960002071 bepotastine Drugs 0.000 description 3
- OQFSQFPPLPISGP-UHFFFAOYSA-N beta-carboxyaspartic acid Natural products OC(=O)C(N)C(C(O)=O)C(O)=O OQFSQFPPLPISGP-UHFFFAOYSA-N 0.000 description 3
- 230000001851 biosynthetic effect Effects 0.000 description 3
- 230000000903 blocking effect Effects 0.000 description 3
- 230000037396 body weight Effects 0.000 description 3
- 210000004899 c-terminal region Anatomy 0.000 description 3
- 125000003178 carboxy group Chemical group [H]OC(*)=O 0.000 description 3
- 230000010261 cell growth Effects 0.000 description 3
- 230000004663 cell proliferation Effects 0.000 description 3
- 210000003850 cellular structure Anatomy 0.000 description 3
- 229950001565 clazakizumab Drugs 0.000 description 3
- 238000003776 cleavage reaction Methods 0.000 description 3
- 230000004540 complement-dependent cytotoxicity Effects 0.000 description 3
- 235000018417 cysteine Nutrition 0.000 description 3
- XUJNEKJLAYXESH-UHFFFAOYSA-N cysteine Natural products SCC(N)C(O)=O XUJNEKJLAYXESH-UHFFFAOYSA-N 0.000 description 3
- 125000000151 cysteine group Chemical group N[C@@H](CS)C(=O)* 0.000 description 3
- 229940127089 cytotoxic agent Drugs 0.000 description 3
- 239000002254 cytotoxic agent Substances 0.000 description 3
- 230000007812 deficiency Effects 0.000 description 3
- 238000001514 detection method Methods 0.000 description 3
- 229910052805 deuterium Inorganic materials 0.000 description 3
- 229940120889 dipyrone Drugs 0.000 description 3
- 238000002845 discoloration Methods 0.000 description 3
- 239000003937 drug carrier Substances 0.000 description 3
- 230000006862 enzymatic digestion Effects 0.000 description 3
- 210000003527 eukaryotic cell Anatomy 0.000 description 3
- 108020005243 folate receptor Proteins 0.000 description 3
- 102000006815 folate receptor Human genes 0.000 description 3
- 235000013922 glutamic acid Nutrition 0.000 description 3
- 239000004220 glutamic acid Substances 0.000 description 3
- HNDVDQJCIGZPNO-UHFFFAOYSA-N histidine Natural products OC(=O)C(N)CC1=CN=CN1 HNDVDQJCIGZPNO-UHFFFAOYSA-N 0.000 description 3
- 235000014304 histidine Nutrition 0.000 description 3
- 210000003701 histiocyte Anatomy 0.000 description 3
- 208000033519 human immunodeficiency virus infectious disease Diseases 0.000 description 3
- 210000002865 immune cell Anatomy 0.000 description 3
- 239000003446 ligand Substances 0.000 description 3
- 210000004698 lymphocyte Anatomy 0.000 description 3
- 238000004949 mass spectrometry Methods 0.000 description 3
- 238000002483 medication Methods 0.000 description 3
- LVWZTYCIRDMTEY-UHFFFAOYSA-N metamizole Chemical compound O=C1C(N(CS(O)(=O)=O)C)=C(C)N(C)N1C1=CC=CC=C1 LVWZTYCIRDMTEY-UHFFFAOYSA-N 0.000 description 3
- 230000000813 microbial effect Effects 0.000 description 3
- 229960005170 moexipril Drugs 0.000 description 3
- 108010059074 monomethylauristatin F Proteins 0.000 description 3
- MQQNFDZXWVTQEH-UHFFFAOYSA-N nafamostat Chemical compound C1=CC(N=C(N)N)=CC=C1C(=O)OC1=CC=C(C=C(C=C2)C(N)=N)C2=C1 MQQNFDZXWVTQEH-UHFFFAOYSA-N 0.000 description 3
- 229950009865 nafamostat Drugs 0.000 description 3
- 210000000440 neutrophil Anatomy 0.000 description 3
- 238000003199 nucleic acid amplification method Methods 0.000 description 3
- 230000008782 phagocytosis Effects 0.000 description 3
- 239000000546 pharmaceutical excipient Substances 0.000 description 3
- 239000000047 product Substances 0.000 description 3
- 230000017854 proteolysis Effects 0.000 description 3
- 230000005180 public health Effects 0.000 description 3
- 238000000746 purification Methods 0.000 description 3
- 238000003127 radioimmunoassay Methods 0.000 description 3
- 230000007017 scission Effects 0.000 description 3
- 238000012216 screening Methods 0.000 description 3
- 238000007423 screening assay Methods 0.000 description 3
- 230000003248 secreting effect Effects 0.000 description 3
- 238000000926 separation method Methods 0.000 description 3
- 150000003384 small molecules Chemical class 0.000 description 3
- 238000004114 suspension culture Methods 0.000 description 3
- 230000009466 transformation Effects 0.000 description 3
- 238000013519 translation Methods 0.000 description 3
- 238000011144 upstream manufacturing Methods 0.000 description 3
- 238000001262 western blot Methods 0.000 description 3
- IAKHMKGGTNLKSZ-INIZCTEOSA-N (S)-colchicine Chemical compound C1([C@@H](NC(C)=O)CC2)=CC(=O)C(OC)=CC=C1C1=C2C=C(OC)C(OC)=C1OC IAKHMKGGTNLKSZ-INIZCTEOSA-N 0.000 description 2
- NFGXHKASABOEEW-UHFFFAOYSA-N 1-methylethyl 11-methoxy-3,7,11-trimethyl-2,4-dodecadienoate Chemical compound COC(C)(C)CCCC(C)CC=CC(C)=CC(=O)OC(C)C NFGXHKASABOEEW-UHFFFAOYSA-N 0.000 description 2
- 208000010470 Ageusia Diseases 0.000 description 2
- 102000009027 Albumins Human genes 0.000 description 2
- 108010088751 Albumins Proteins 0.000 description 2
- 206010002653 Anosmia Diseases 0.000 description 2
- 108090001008 Avidin Proteins 0.000 description 2
- 241000112287 Bat coronavirus Species 0.000 description 2
- 241000008904 Betacoronavirus Species 0.000 description 2
- 206010006187 Breast cancer Diseases 0.000 description 2
- 241000282465 Canis Species 0.000 description 2
- 241000282552 Chlorocebus aethiops Species 0.000 description 2
- 208000006545 Chronic Obstructive Pulmonary Disease Diseases 0.000 description 2
- 108020004705 Codon Proteins 0.000 description 2
- 108020004635 Complementary DNA Proteins 0.000 description 2
- TURXCFUGBPBPRS-UHFFFAOYSA-N Curvulin Chemical compound CCOC(=O)CC1=CC(O)=CC(O)=C1C(C)=O TURXCFUGBPBPRS-UHFFFAOYSA-N 0.000 description 2
- 206010050685 Cytokine storm Diseases 0.000 description 2
- 102000053602 DNA Human genes 0.000 description 2
- 206010012735 Diarrhoea Diseases 0.000 description 2
- 108010067722 Dipeptidyl Peptidase 4 Proteins 0.000 description 2
- AOJJSUZBOXZQNB-TZSSRYMLSA-N Doxorubicin Chemical compound O([C@H]1C[C@@](O)(CC=2C(O)=C3C(=O)C=4C=CC=C(C=4C(=O)C3=C(O)C=21)OC)C(=O)CO)[C@H]1C[C@H](N)[C@H](O)[C@H](C)O1 AOJJSUZBOXZQNB-TZSSRYMLSA-N 0.000 description 2
- 206010059866 Drug resistance Diseases 0.000 description 2
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 description 2
- 241000196324 Embryophyta Species 0.000 description 2
- YQYJSBFKSSDGFO-UHFFFAOYSA-N Epihygromycin Natural products OC1C(O)C(C(=O)C)OC1OC(C(=C1)O)=CC=C1C=C(C)C(=O)NC1C(O)C(O)C2OCOC2C1O YQYJSBFKSSDGFO-UHFFFAOYSA-N 0.000 description 2
- 241000588724 Escherichia coli Species 0.000 description 2
- 241000206602 Eukaryota Species 0.000 description 2
- 108091029865 Exogenous DNA Proteins 0.000 description 2
- WQZGKKKJIJFFOK-GASJEMHNSA-N Glucose Natural products OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 2
- 208000032843 Hemorrhage Diseases 0.000 description 2
- 101000878602 Homo sapiens Immunoglobulin alpha Fc receptor Proteins 0.000 description 2
- 108010073816 IgE Receptors Proteins 0.000 description 2
- 102000009438 IgE Receptors Human genes 0.000 description 2
- 108010073807 IgG Receptors Proteins 0.000 description 2
- 102000009490 IgG Receptors Human genes 0.000 description 2
- 102000006496 Immunoglobulin Heavy Chains Human genes 0.000 description 2
- 108010019476 Immunoglobulin Heavy Chains Proteins 0.000 description 2
- 102100038005 Immunoglobulin alpha Fc receptor Human genes 0.000 description 2
- 102100023915 Insulin Human genes 0.000 description 2
- 108090001061 Insulin Proteins 0.000 description 2
- 208000029523 Interstitial Lung disease Diseases 0.000 description 2
- 229940024452 Janssen COVID-19 vaccine Drugs 0.000 description 2
- XUJNEKJLAYXESH-REOHCLBHSA-N L-Cysteine Chemical compound SC[C@H](N)C(O)=O XUJNEKJLAYXESH-REOHCLBHSA-N 0.000 description 2
- 238000000585 Mann–Whitney U test Methods 0.000 description 2
- 241000699673 Mesocricetus auratus Species 0.000 description 2
- 102000003792 Metallothionein Human genes 0.000 description 2
- NWIBSHFKIJFRCO-WUDYKRTCSA-N Mytomycin Chemical compound C1N2C(C(C(C)=C(N)C3=O)=O)=C3[C@@H](COC(N)=O)[C@@]2(OC)[C@@H]2[C@H]1N2 NWIBSHFKIJFRCO-WUDYKRTCSA-N 0.000 description 2
- 206010028980 Neoplasm Diseases 0.000 description 2
- 206010030113 Oedema Diseases 0.000 description 2
- 108091034117 Oligonucleotide Proteins 0.000 description 2
- 241000283973 Oryctolagus cuniculus Species 0.000 description 2
- 208000002193 Pain Diseases 0.000 description 2
- 108090000526 Papain Proteins 0.000 description 2
- KPKZJLCSROULON-QKGLWVMZSA-N Phalloidin Chemical compound N1C(=O)[C@@H]([C@@H](O)C)NC(=O)[C@H](C)NC(=O)[C@H](C[C@@](C)(O)CO)NC(=O)[C@H](C2)NC(=O)[C@H](C)NC(=O)[C@@H]3C[C@H](O)CN3C(=O)[C@@H]1CSC1=C2C2=CC=CC=C2N1 KPKZJLCSROULON-QKGLWVMZSA-N 0.000 description 2
- 206010035226 Plasma cell myeloma Diseases 0.000 description 2
- 241000288906 Primates Species 0.000 description 2
- 229940125677 REGEN-COV Drugs 0.000 description 2
- 239000012980 RPMI-1640 medium Substances 0.000 description 2
- 241000700157 Rattus norvegicus Species 0.000 description 2
- 108020005091 Replication Origin Proteins 0.000 description 2
- 208000036071 Rhinorrhea Diseases 0.000 description 2
- 206010039101 Rhinorrhoea Diseases 0.000 description 2
- 241000283984 Rodentia Species 0.000 description 2
- INVGWHRKADIJHF-UHFFFAOYSA-N Sanguinarin Chemical compound C1=C2OCOC2=CC2=C3[N+](C)=CC4=C(OCO5)C5=CC=C4C3=CC=C21 INVGWHRKADIJHF-UHFFFAOYSA-N 0.000 description 2
- 206010040047 Sepsis Diseases 0.000 description 2
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 2
- 108010090804 Streptavidin Proteins 0.000 description 2
- 238000000692 Student's t-test Methods 0.000 description 2
- 210000001744 T-lymphocyte Anatomy 0.000 description 2
- 108010022394 Threonine synthase Proteins 0.000 description 2
- IQFYYKKMVGJFEH-XLPZGREQSA-N Thymidine Chemical compound O=C1NC(=O)C(C)=CN1[C@@H]1O[C@H](CO)[C@@H](O)C1 IQFYYKKMVGJFEH-XLPZGREQSA-N 0.000 description 2
- 102000006601 Thymidine Kinase Human genes 0.000 description 2
- 108020004440 Thymidine kinase Proteins 0.000 description 2
- 102000004338 Transferrin Human genes 0.000 description 2
- 108090000901 Transferrin Proteins 0.000 description 2
- 241000711484 Transmissible gastroenteritis virus Species 0.000 description 2
- 108091023045 Untranslated Region Proteins 0.000 description 2
- 244000000188 Vaccinium ovalifolium Species 0.000 description 2
- 241000711975 Vesicular stomatitis virus Species 0.000 description 2
- 206010058874 Viraemia Diseases 0.000 description 2
- 108020005202 Viral DNA Proteins 0.000 description 2
- 108020000999 Viral RNA Proteins 0.000 description 2
- IEDXPSOJFSVCKU-HOKPPMCLSA-N [4-[[(2S)-5-(carbamoylamino)-2-[[(2S)-2-[6-(2,5-dioxopyrrolidin-1-yl)hexanoylamino]-3-methylbutanoyl]amino]pentanoyl]amino]phenyl]methyl N-[(2S)-1-[[(2S)-1-[[(3R,4S,5S)-1-[(2S)-2-[(1R,2R)-3-[[(1S,2R)-1-hydroxy-1-phenylpropan-2-yl]amino]-1-methoxy-2-methyl-3-oxopropyl]pyrrolidin-1-yl]-3-methoxy-5-methyl-1-oxoheptan-4-yl]-methylamino]-3-methyl-1-oxobutan-2-yl]amino]-3-methyl-1-oxobutan-2-yl]-N-methylcarbamate Chemical compound CC[C@H](C)[C@@H]([C@@H](CC(=O)N1CCC[C@H]1[C@H](OC)[C@@H](C)C(=O)N[C@H](C)[C@@H](O)c1ccccc1)OC)N(C)C(=O)[C@@H](NC(=O)[C@H](C(C)C)N(C)C(=O)OCc1ccc(NC(=O)[C@H](CCCNC(N)=O)NC(=O)[C@@H](NC(=O)CCCCCN2C(=O)CCC2=O)C(C)C)cc1)C(C)C IEDXPSOJFSVCKU-HOKPPMCLSA-N 0.000 description 2
- 239000013543 active substance Substances 0.000 description 2
- OIRDTQYFTABQOQ-KQYNXXCUSA-N adenosine Chemical compound C1=NC=2C(N)=NC=NC=2N1[C@@H]1O[C@H](CO)[C@@H](O)[C@H]1O OIRDTQYFTABQOQ-KQYNXXCUSA-N 0.000 description 2
- 235000019666 ageusia Nutrition 0.000 description 2
- 125000003295 alanine group Chemical group N[C@@H](C)C(=O)* 0.000 description 2
- 210000002588 alveolar type II cell Anatomy 0.000 description 2
- 125000003277 amino group Chemical group 0.000 description 2
- 229960000723 ampicillin Drugs 0.000 description 2
- 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 2
- 229940121363 anti-inflammatory agent Drugs 0.000 description 2
- 230000002141 anti-parasite Effects 0.000 description 2
- 229940124350 antibacterial drug Drugs 0.000 description 2
- 230000005888 antibody-dependent cellular phagocytosis Effects 0.000 description 2
- QVGXLLKOCUKJST-UHFFFAOYSA-N atomic oxygen Chemical compound [O] QVGXLLKOCUKJST-UHFFFAOYSA-N 0.000 description 2
- WQZGKKKJIJFFOK-VFUOTHLCSA-N beta-D-glucose Chemical compound OC[C@H]1O[C@@H](O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-VFUOTHLCSA-N 0.000 description 2
- 230000015572 biosynthetic process Effects 0.000 description 2
- 229960002685 biotin Drugs 0.000 description 2
- 235000020958 biotin Nutrition 0.000 description 2
- 239000011616 biotin Substances 0.000 description 2
- 210000004204 blood vessel Anatomy 0.000 description 2
- 210000004556 brain Anatomy 0.000 description 2
- 230000001413 cellular effect Effects 0.000 description 2
- 208000019065 cervical carcinoma Diseases 0.000 description 2
- 238000004587 chromatography analysis Methods 0.000 description 2
- 238000005352 clarification Methods 0.000 description 2
- 208000027744 congestion Diseases 0.000 description 2
- 238000007596 consolidation process Methods 0.000 description 2
- 230000001276 controlling effect Effects 0.000 description 2
- 210000004748 cultured cell Anatomy 0.000 description 2
- 231100000599 cytotoxic agent Toxicity 0.000 description 2
- 238000010586 diagram Methods 0.000 description 2
- 102000004419 dihydrofolate reductase Human genes 0.000 description 2
- 238000010494 dissociation reaction Methods 0.000 description 2
- 230000005593 dissociations Effects 0.000 description 2
- 238000005516 engineering process Methods 0.000 description 2
- 230000002708 enhancing effect Effects 0.000 description 2
- 238000000855 fermentation Methods 0.000 description 2
- 230000004151 fermentation Effects 0.000 description 2
- 239000007850 fluorescent dye Substances 0.000 description 2
- 230000002068 genetic effect Effects 0.000 description 2
- 239000008103 glucose Substances 0.000 description 2
- 230000013595 glycosylation Effects 0.000 description 2
- 238000006206 glycosylation reaction Methods 0.000 description 2
- 238000002744 homologous recombination Methods 0.000 description 2
- 230000006801 homologous recombination Effects 0.000 description 2
- 230000005099 host tropism Effects 0.000 description 2
- 230000002209 hydrophobic effect Effects 0.000 description 2
- 229910052588 hydroxylapatite Inorganic materials 0.000 description 2
- 230000002163 immunogen Effects 0.000 description 2
- 230000005847 immunogenicity Effects 0.000 description 2
- 230000016784 immunoglobulin production Effects 0.000 description 2
- 230000006872 improvement Effects 0.000 description 2
- 239000007924 injection Substances 0.000 description 2
- 238000002347 injection Methods 0.000 description 2
- 229940125396 insulin Drugs 0.000 description 2
- 239000007927 intramuscular injection Substances 0.000 description 2
- 210000003734 kidney Anatomy 0.000 description 2
- 238000012417 linear regression Methods 0.000 description 2
- 210000005265 lung cell Anatomy 0.000 description 2
- 125000003588 lysine group Chemical group [H]N([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])(N([H])[H])C(*)=O 0.000 description 2
- 239000012528 membrane Substances 0.000 description 2
- 108020004999 messenger RNA Proteins 0.000 description 2
- MYWUZJCMWCOHBA-VIFPVBQESA-N methamphetamine Chemical compound CN[C@@H](C)CC1=CC=CC=C1 MYWUZJCMWCOHBA-VIFPVBQESA-N 0.000 description 2
- 125000002496 methyl group Chemical group [H]C([H])([H])* 0.000 description 2
- 108010093470 monomethyl auristatin E Proteins 0.000 description 2
- DNIAPMSPPWPWGF-UHFFFAOYSA-N monopropylene glycol Natural products CC(O)CO DNIAPMSPPWPWGF-UHFFFAOYSA-N 0.000 description 2
- 201000000050 myeloid neoplasm Diseases 0.000 description 2
- 230000007935 neutral effect Effects 0.000 description 2
- 238000005457 optimization Methods 0.000 description 2
- 210000001672 ovary Anatomy 0.000 description 2
- 230000003647 oxidation Effects 0.000 description 2
- 238000007254 oxidation reaction Methods 0.000 description 2
- 229910052760 oxygen Inorganic materials 0.000 description 2
- 239000001301 oxygen Substances 0.000 description 2
- 230000020477 pH reduction Effects 0.000 description 2
- 229940055729 papain Drugs 0.000 description 2
- 235000019834 papain Nutrition 0.000 description 2
- 230000001575 pathological effect Effects 0.000 description 2
- 230000007170 pathology Effects 0.000 description 2
- XYJRXVWERLGGKC-UHFFFAOYSA-D pentacalcium;hydroxide;triphosphate Chemical compound [OH-].[Ca+2].[Ca+2].[Ca+2].[Ca+2].[Ca+2].[O-]P([O-])([O-])=O.[O-]P([O-])([O-])=O.[O-]P([O-])([O-])=O XYJRXVWERLGGKC-UHFFFAOYSA-D 0.000 description 2
- 230000008884 pinocytosis Effects 0.000 description 2
- 210000002381 plasma Anatomy 0.000 description 2
- 230000008488 polyadenylation Effects 0.000 description 2
- 230000002035 prolonged effect Effects 0.000 description 2
- 235000019419 proteases Nutrition 0.000 description 2
- 230000001681 protective effect Effects 0.000 description 2
- 238000000159 protein binding assay Methods 0.000 description 2
- RXWNCPJZOCPEPQ-NVWDDTSBSA-N puromycin Chemical compound C1=CC(OC)=CC=C1C[C@H](N)C(=O)N[C@H]1[C@@H](O)[C@H](N2C3=NC=NC(=C3N=C2)N(C)C)O[C@@H]1CO RXWNCPJZOCPEPQ-NVWDDTSBSA-N 0.000 description 2
- 230000009257 reactivity Effects 0.000 description 2
- 208000023504 respiratory system disease Diseases 0.000 description 2
- 238000009738 saturating Methods 0.000 description 2
- 238000004088 simulation Methods 0.000 description 2
- LMXOHSDXUQEUSF-YECHIGJVSA-N sinefungin Chemical compound O[C@@H]1[C@H](O)[C@@H](C[C@H](CC[C@H](N)C(O)=O)N)O[C@H]1N1C2=NC=NC(N)=C2N=C1 LMXOHSDXUQEUSF-YECHIGJVSA-N 0.000 description 2
- 230000009870 specific binding Effects 0.000 description 2
- 230000000087 stabilizing effect Effects 0.000 description 2
- 230000035882 stress Effects 0.000 description 2
- 239000000758 substrate Substances 0.000 description 2
- 230000002194 synthesizing effect Effects 0.000 description 2
- 238000012353 t test Methods 0.000 description 2
- 229940124597 therapeutic agent Drugs 0.000 description 2
- 230000002103 transcriptional effect Effects 0.000 description 2
- 239000012581 transferrin Substances 0.000 description 2
- 230000029812 viral genome replication Effects 0.000 description 2
- 238000005406 washing Methods 0.000 description 2
- NGGMYCMLYOUNGM-UHFFFAOYSA-N (-)-fumagillin Natural products O1C(CC=C(C)C)C1(C)C1C(OC)C(OC(=O)C=CC=CC=CC=CC(O)=O)CCC21CO2 NGGMYCMLYOUNGM-UHFFFAOYSA-N 0.000 description 1
- MNULEGDCPYONBU-WMBHJXFZSA-N (1r,4s,5e,5'r,6'r,7e,10s,11r,12s,14r,15s,16s,18r,19s,20r,21e,25s,26r,27s,29s)-4-ethyl-11,12,15,19-tetrahydroxy-6'-[(2s)-2-hydroxypropyl]-5',10,12,14,16,18,20,26,29-nonamethylspiro[24,28-dioxabicyclo[23.3.1]nonacosa-5,7,21-triene-27,2'-oxane]-13,17,23-trio Polymers O([C@@H]1CC[C@@H](/C=C/C=C/C[C@H](C)[C@@H](O)[C@](C)(O)C(=O)[C@H](C)[C@@H](O)[C@H](C)C(=O)[C@H](C)[C@@H](O)[C@H](C)/C=C/C(=O)O[C@H]([C@H]2C)[C@H]1C)CC)[C@]12CC[C@@H](C)[C@@H](C[C@H](C)O)O1 MNULEGDCPYONBU-WMBHJXFZSA-N 0.000 description 1
- MNULEGDCPYONBU-DJRUDOHVSA-N (1s,4r,5z,5'r,6'r,7e,10s,11r,12s,14r,15s,18r,19r,20s,21e,26r,27s)-4-ethyl-11,12,15,19-tetrahydroxy-6'-(2-hydroxypropyl)-5',10,12,14,16,18,20,26,29-nonamethylspiro[24,28-dioxabicyclo[23.3.1]nonacosa-5,7,21-triene-27,2'-oxane]-13,17,23-trione Polymers O([C@H]1CC[C@H](\C=C/C=C/C[C@H](C)[C@@H](O)[C@](C)(O)C(=O)[C@H](C)[C@@H](O)C(C)C(=O)[C@H](C)[C@H](O)[C@@H](C)/C=C/C(=O)OC([C@H]2C)C1C)CC)[C@]12CC[C@@H](C)[C@@H](CC(C)O)O1 MNULEGDCPYONBU-DJRUDOHVSA-N 0.000 description 1
- BLUGYPPOFIHFJS-UUFHNPECSA-N (2s)-n-[(2s)-1-[[(3r,4s,5s)-3-methoxy-1-[(2s)-2-[(1r,2r)-1-methoxy-2-methyl-3-oxo-3-[[(1s)-2-phenyl-1-(1,3-thiazol-2-yl)ethyl]amino]propyl]pyrrolidin-1-yl]-5-methyl-1-oxoheptan-4-yl]-methylamino]-3-methyl-1-oxobutan-2-yl]-3-methyl-2-(methylamino)butanamid Chemical compound CN[C@@H](C(C)C)C(=O)N[C@@H](C(C)C)C(=O)N(C)[C@@H]([C@@H](C)CC)[C@H](OC)CC(=O)N1CCC[C@H]1[C@H](OC)[C@@H](C)C(=O)N[C@H](C=1SC=CN=1)CC1=CC=CC=C1 BLUGYPPOFIHFJS-UUFHNPECSA-N 0.000 description 1
- DLKUYSQUHXBYPB-NSSHGSRYSA-N (2s,4r)-4-[[2-[(1r,3r)-1-acetyloxy-4-methyl-3-[3-methylbutanoyloxymethyl-[(2s,3s)-3-methyl-2-[[(2r)-1-methylpiperidine-2-carbonyl]amino]pentanoyl]amino]pentyl]-1,3-thiazole-4-carbonyl]amino]-2-methyl-5-(4-methylphenyl)pentanoic acid Chemical compound N([C@@H]([C@@H](C)CC)C(=O)N(COC(=O)CC(C)C)[C@H](C[C@@H](OC(C)=O)C=1SC=C(N=1)C(=O)N[C@H](C[C@H](C)C(O)=O)CC=1C=CC(C)=CC=1)C(C)C)C(=O)[C@H]1CCCCN1C DLKUYSQUHXBYPB-NSSHGSRYSA-N 0.000 description 1
- OMKSBDLWMROKNU-XYOKQWHBSA-N (3e)-3-[(2,5-dihydroxyphenyl)methylidene]-1h-indol-2-one Chemical compound OC1=CC=C(O)C(\C=C\2C3=CC=CC=C3NC/2=O)=C1 OMKSBDLWMROKNU-XYOKQWHBSA-N 0.000 description 1
- WILMROCKORZEMQ-AIUMZUNXSA-N (3e,5e,7e,9r,10r,11e,13e,17s,18s,20s)-20-[(r)-[(2r,3r,4s,5r,6r)-2,4-dihydroxy-6-[(2r)-2-[(2r,4s,5s,6s)-4-hydroxy-5-[(2s,4r,5r,6r)-5-hydroxy-4-methoxy-6-methyloxan-2-yl]oxy-4,6-dimethyloxan-2-yl]oxy-3-methoxypropyl]-3,5-dimethyloxan-2-yl]-hydroxymethyl]-10 Chemical compound O([C@@H]1/C=C/C(/C)=C/CC[C@H](O)[C@@H](OC)C[C@H](OC(=O)/C(C)=C/C(/C)=C/C(/C)=C/[C@H]1C)[C@@H](O)[C@@]1(O[C@@H]([C@@H]([C@H](O)[C@H]1C)C)C[C@H](COC)O[C@@H]1O[C@@H](C)[C@H](O[C@@H]2O[C@H](C)[C@@H](O)[C@H](OC)C2)[C@@](C)(O)C1)O)[C@@H]1O[C@@H](C)[C@H](OC)[C@@H](O)[C@@H]1O WILMROCKORZEMQ-AIUMZUNXSA-N 0.000 description 1
- SGYJGGKDGBXCNY-QXUYBEEESA-N (3s,9s,12r)-3-benzyl-6,6-dimethyl-9-[6-[(2s)-oxiran-2-yl]-6-oxohexyl]-1,4,7,10-tetrazabicyclo[10.3.0]pentadecane-2,5,8,11-tetrone Chemical compound C([C@H]1C(=O)NC(C(N[C@@H](CC=2C=CC=CC=2)C(=O)N2CCC[C@@H]2C(=O)N1)=O)(C)C)CCCCC(=O)[C@@H]1CO1 SGYJGGKDGBXCNY-QXUYBEEESA-N 0.000 description 1
- MNULEGDCPYONBU-YNZHUHFTSA-N (4Z,18Z,20Z)-22-ethyl-7,11,14,15-tetrahydroxy-6'-(2-hydroxypropyl)-5',6,8,10,12,14,16,28,29-nonamethylspiro[2,26-dioxabicyclo[23.3.1]nonacosa-4,18,20-triene-27,2'-oxane]-3,9,13-trione Polymers CC1C(C2C)OC(=O)\C=C/C(C)C(O)C(C)C(=O)C(C)C(O)C(C)C(=O)C(C)(O)C(O)C(C)C\C=C/C=C\C(CC)CCC2OC21CCC(C)C(CC(C)O)O2 MNULEGDCPYONBU-YNZHUHFTSA-N 0.000 description 1
- MNULEGDCPYONBU-VVXVDZGXSA-N (5e,5'r,7e,10s,11r,12s,14s,15r,16r,18r,19s,20r,21e,26r,29s)-4-ethyl-11,12,15,19-tetrahydroxy-6'-[(2s)-2-hydroxypropyl]-5',10,12,14,16,18,20,26,29-nonamethylspiro[24,28-dioxabicyclo[23.3.1]nonacosa-5,7,21-triene-27,2'-oxane]-13,17,23-trione Polymers C([C@H](C)[C@@H](O)[C@](C)(O)C(=O)[C@@H](C)[C@H](O)[C@@H](C)C(=O)[C@H](C)[C@@H](O)[C@H](C)/C=C/C(=O)OC([C@H]1C)[C@H]2C)\C=C\C=C\C(CC)CCC2OC21CC[C@@H](C)C(C[C@H](C)O)O2 MNULEGDCPYONBU-VVXVDZGXSA-N 0.000 description 1
- CNTMOLDWXSVYKD-PSRNMDMQSA-N (e,4s)-4-[[(2s)-3,3-dimethyl-2-[[(2s)-3-methyl-2-(methylamino)-3-phenylbutanoyl]amino]butanoyl]-methylamino]-2,5-dimethylhex-2-enoic acid Chemical compound OC(=O)C(/C)=C/[C@H](C(C)C)N(C)C(=O)[C@H](C(C)(C)C)NC(=O)[C@@H](NC)C(C)(C)C1=CC=CC=C1 CNTMOLDWXSVYKD-PSRNMDMQSA-N 0.000 description 1
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 1
- MPGIDXBHXRMMSY-UHFFFAOYSA-N 1-(4,6-dipentyl-5,6-dihydrooxadiazin-2-yl)-3-hydroxypropan-1-one Chemical compound C(CCCC)C1=NN(OC(C1)CCCCC)C(CCO)=O MPGIDXBHXRMMSY-UHFFFAOYSA-N 0.000 description 1
- CXCHEKCRJQRVNG-UHFFFAOYSA-N 2,2,2-trifluoroethanesulfonyl chloride Chemical compound FC(F)(F)CS(Cl)(=O)=O CXCHEKCRJQRVNG-UHFFFAOYSA-N 0.000 description 1
- ISZXEMUWHQLLTC-LSIVYLFASA-N 2-[(2r,5s,6s)-6-[(2e,4e,6s)-7-[(2r,3r)-3-[(2r,3r,4r)-4-hydroxy-3-methoxypentan-2-yl]-2-methyloxiran-2-yl]-6-methylhepta-2,4-dien-2-yl]-5-methyloxan-2-yl]acetic acid Chemical compound CO[C@@H]([C@@H](C)O)[C@@H](C)[C@H]1O[C@]1(C)C[C@H](C)\C=C\C=C(/C)[C@@H]1[C@@H](C)CC[C@H](CC(O)=O)O1 ISZXEMUWHQLLTC-LSIVYLFASA-N 0.000 description 1
- JKMHFZQWWAIEOD-UHFFFAOYSA-N 2-[4-(2-hydroxyethyl)piperazin-1-yl]ethanesulfonic acid Chemical compound OCC[NH+]1CCN(CCS([O-])(=O)=O)CC1 JKMHFZQWWAIEOD-UHFFFAOYSA-N 0.000 description 1
- IDCOTQWQVPRTNK-UHFFFAOYSA-N 2-n,6-n-bis[(4-methoxyphenyl)methyl]-9-propan-2-ylpurine-2,6-diamine Chemical compound C1=CC(OC)=CC=C1CNC1=NC(NCC=2C=CC(OC)=CC=2)=C(N=CN2C(C)C)C2=N1 IDCOTQWQVPRTNK-UHFFFAOYSA-N 0.000 description 1
- 238000005084 2D-nuclear magnetic resonance Methods 0.000 description 1
- MNULEGDCPYONBU-UHFFFAOYSA-N 4-ethyl-11,12,15,19-tetrahydroxy-6'-(2-hydroxypropyl)-5',10,12,14,16,18,20,26,29-nonamethylspiro[24,28-dioxabicyclo[23.3.1]nonacosa-5,7,21-triene-27,2'-oxane]-13,17,23-trione Polymers CC1C(C2C)OC(=O)C=CC(C)C(O)C(C)C(=O)C(C)C(O)C(C)C(=O)C(C)(O)C(O)C(C)CC=CC=CC(CC)CCC2OC21CCC(C)C(CC(C)O)O2 MNULEGDCPYONBU-UHFFFAOYSA-N 0.000 description 1
- 101710169336 5'-deoxyadenosine deaminase Proteins 0.000 description 1
- LGZKGOGODCLQHG-CYBMUJFWSA-N 5-[(2r)-2-hydroxy-2-(3,4,5-trimethoxyphenyl)ethyl]-2-methoxyphenol Chemical compound C1=C(O)C(OC)=CC=C1C[C@@H](O)C1=CC(OC)=C(OC)C(OC)=C1 LGZKGOGODCLQHG-CYBMUJFWSA-N 0.000 description 1
- STQGQHZAVUOBTE-UHFFFAOYSA-N 7-Cyan-hept-2t-en-4,6-diinsaeure Natural products C1=2C(O)=C3C(=O)C=4C(OC)=CC=CC=4C(=O)C3=C(O)C=2CC(O)(C(C)=O)CC1OC1CC(N)C(O)C(C)O1 STQGQHZAVUOBTE-UHFFFAOYSA-N 0.000 description 1
- FJHBVJOVLFPMQE-QFIPXVFZSA-N 7-Ethyl-10-Hydroxy-Camptothecin Chemical compound C1=C(O)C=C2C(CC)=C(CN3C(C4=C([C@@](C(=O)OC4)(O)CC)C=C33)=O)C3=NC2=C1 FJHBVJOVLFPMQE-QFIPXVFZSA-N 0.000 description 1
- CJIJXIFQYOPWTF-UHFFFAOYSA-N 7-hydroxycoumarin Natural products O1C(=O)C=CC2=CC(O)=CC=C21 CJIJXIFQYOPWTF-UHFFFAOYSA-N 0.000 description 1
- LPXQRXLUHJKZIE-UHFFFAOYSA-N 8-azaguanine Chemical compound NC1=NC(O)=C2NN=NC2=N1 LPXQRXLUHJKZIE-UHFFFAOYSA-N 0.000 description 1
- OZOGDCZJYVSUBR-UHFFFAOYSA-N 8-chloro-n-[4-(trifluoromethoxy)phenyl]quinolin-2-amine Chemical compound C1=CC(OC(F)(F)F)=CC=C1NC1=CC=C(C=CC=C2Cl)C2=N1 OZOGDCZJYVSUBR-UHFFFAOYSA-N 0.000 description 1
- QZTWUDDGLIDXSE-UHFFFAOYSA-N 9-hydroxyellipticine Chemical compound N1=CC=C2C(C)=C(NC=3C4=CC(O)=CC=3)C4=C(C)C2=C1 QZTWUDDGLIDXSE-UHFFFAOYSA-N 0.000 description 1
- 208000007934 ACTH-independent macronodular adrenal hyperplasia Diseases 0.000 description 1
- 208000030507 AIDS Diseases 0.000 description 1
- 102000012440 Acetylcholinesterase Human genes 0.000 description 1
- 108010022752 Acetylcholinesterase Proteins 0.000 description 1
- 102000007469 Actins Human genes 0.000 description 1
- 108010085238 Actins Proteins 0.000 description 1
- 102000055025 Adenosine deaminases Human genes 0.000 description 1
- 108010000239 Aequorin Proteins 0.000 description 1
- BGYNLOSBKBOJJD-IUCAKERBSA-N Aeroplysinin 1 Chemical compound COC1=C(Br)[C@H](O)[C@](O)(CC#N)C=C1Br BGYNLOSBKBOJJD-IUCAKERBSA-N 0.000 description 1
- ZGCSNRKSJLVANE-UHFFFAOYSA-N Aglycone-Rebeccamycin Natural products N1C2=C3NC4=C(Cl)C=CC=C4C3=C(C(=O)NC3=O)C3=C2C2=C1C(Cl)=CC=C2 ZGCSNRKSJLVANE-UHFFFAOYSA-N 0.000 description 1
- 102000002260 Alkaline Phosphatase Human genes 0.000 description 1
- 108020004774 Alkaline Phosphatase Proteins 0.000 description 1
- 102100023635 Alpha-fetoprotein Human genes 0.000 description 1
- 108010027164 Amanitins Proteins 0.000 description 1
- 101710154825 Aminoglycoside 3'-phosphotransferase Proteins 0.000 description 1
- 101100155017 Arabidopsis thaliana RCE2 gene Proteins 0.000 description 1
- 208000023275 Autoimmune disease Diseases 0.000 description 1
- 241000713842 Avian sarcoma virus Species 0.000 description 1
- 108091008875 B cell receptors Proteins 0.000 description 1
- 241000304886 Bacilli Species 0.000 description 1
- 241000207900 Bat Coronavirus Rhhar/CII_KSA_004/Bisha/Saudi Arabia/2013 Species 0.000 description 1
- 241000207916 Bat coronavirus Pikuh/CII_KSA_001/Riyadh/Saudi Arabia/2013 Species 0.000 description 1
- 241000207918 Bat coronavirus Rhhar/CII_KSA_002/Bisha/Saudi Arabia/2013 Species 0.000 description 1
- 241000207920 Bat coronavirus Rhhar/CII_KSA_003/Bisha/Saudi Arabia/2013 Species 0.000 description 1
- DWRXFEITVBNRMK-UHFFFAOYSA-N Beta-D-1-Arabinofuranosylthymine Natural products O=C1NC(=O)C(C)=CN1C1C(O)C(O)C(CO)O1 DWRXFEITVBNRMK-UHFFFAOYSA-N 0.000 description 1
- 102000004506 Blood Proteins Human genes 0.000 description 1
- 108010017384 Blood Proteins Proteins 0.000 description 1
- 241000283690 Bos taurus Species 0.000 description 1
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 1
- 241000711443 Bovine coronavirus Species 0.000 description 1
- 241000701822 Bovine papillomavirus Species 0.000 description 1
- 241000701922 Bovine parvovirus Species 0.000 description 1
- 240000007124 Brassica oleracea Species 0.000 description 1
- 206010006458 Bronchitis chronic Diseases 0.000 description 1
- 125000001433 C-terminal amino-acid group Chemical group 0.000 description 1
- 239000002126 C01EB10 - Adenosine Substances 0.000 description 1
- 238000011357 CAR T-cell therapy Methods 0.000 description 1
- 101100074330 Caenorhabditis elegans lec-8 gene Proteins 0.000 description 1
- OYPRJOBELJOOCE-UHFFFAOYSA-N Calcium Chemical compound [Ca] OYPRJOBELJOOCE-UHFFFAOYSA-N 0.000 description 1
- OKTJSMMVPCPJKN-NJFSPNSNSA-N Carbon-14 Chemical compound [14C] OKTJSMMVPCPJKN-NJFSPNSNSA-N 0.000 description 1
- 229920002134 Carboxymethyl cellulose Polymers 0.000 description 1
- 208000031229 Cardiomyopathies Diseases 0.000 description 1
- 102000000844 Cell Surface Receptors Human genes 0.000 description 1
- 108010001857 Cell Surface Receptors Proteins 0.000 description 1
- 108050001186 Chaperonin Cpn60 Proteins 0.000 description 1
- 102000052603 Chaperonins Human genes 0.000 description 1
- 206010008479 Chest Pain Diseases 0.000 description 1
- 241000288673 Chiroptera Species 0.000 description 1
- SGYJGGKDGBXCNY-UHFFFAOYSA-N Chlamydocin Natural products N1C(=O)C2CCCN2C(=O)C(CC=2C=CC=CC=2)NC(=O)C(C)(C)NC(=O)C1CCCCCC(=O)C1CO1 SGYJGGKDGBXCNY-UHFFFAOYSA-N 0.000 description 1
- 241000251730 Chondrichthyes Species 0.000 description 1
- 108090000317 Chymotrypsin Proteins 0.000 description 1
- 108091062157 Cis-regulatory element Proteins 0.000 description 1
- 101100007328 Cocos nucifera COS-1 gene Proteins 0.000 description 1
- 208000035473 Communicable disease Diseases 0.000 description 1
- 208000002330 Congenital Heart Defects Diseases 0.000 description 1
- KQLDDLUWUFBQHP-UHFFFAOYSA-N Cordycepin Natural products C1=NC=2C(N)=NC=NC=2N1C1OCC(CO)C1O KQLDDLUWUFBQHP-UHFFFAOYSA-N 0.000 description 1
- 108010061994 Coronavirus Spike Glycoprotein Proteins 0.000 description 1
- 241000723655 Cowpea mosaic virus Species 0.000 description 1
- LNSXRXFBSDRILE-UHFFFAOYSA-N Cucurbitacin Natural products CC(=O)OC(C)(C)C=CC(=O)C(C)(O)C1C(O)CC2(C)C3CC=C4C(C)(C)C(O)C(O)CC4(C)C3(C)C(=O)CC12C LNSXRXFBSDRILE-UHFFFAOYSA-N 0.000 description 1
- CVKKIVYBGGDJCR-SXDZHWHFSA-N Cucurbitacin B Natural products CC(=O)OC(C)(C)C=CC(=O)[C@@](C)(O)[C@@H]1[C@@H](O)C[C@]2(C)C3=CC[C@@H]4C(C)(C)C(=O)[C@H](O)C[C@@]4(C)[C@@H]3CC(=O)[C@@]12C CVKKIVYBGGDJCR-SXDZHWHFSA-N 0.000 description 1
- SRUWWOSWHXIIIA-UKPGNTDSSA-N Cyanoginosin Chemical compound N1C(=O)[C@H](CCCN=C(N)N)NC(=O)[C@@H](C)[C@H](C(O)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](C)NC(=O)C(=C)N(C)C(=O)CC[C@H](C(O)=O)N(C)C(=O)[C@@H](C)[C@@H]1\C=C\C(\C)=C\[C@H](C)[C@@H](O)CC1=CC=CC=C1 SRUWWOSWHXIIIA-UKPGNTDSSA-N 0.000 description 1
- QASFUMOKHFSJGL-LAFRSMQTSA-N Cyclopamine Chemical compound C1C=C2C[C@@H](O)CC[C@]2(C)[C@@H](CC2=C3C)[C@@H]1[C@@H]2CC[C@@]13O[C@@H]2C[C@H](C)CN[C@H]2[C@H]1C QASFUMOKHFSJGL-LAFRSMQTSA-N 0.000 description 1
- 102000001493 Cyclophilins Human genes 0.000 description 1
- 108010068682 Cyclophilins Proteins 0.000 description 1
- 101710112752 Cytotoxin Proteins 0.000 description 1
- IGXWBGJHJZYPQS-SSDOTTSWSA-N D-Luciferin Chemical compound OC(=O)[C@H]1CSC(C=2SC3=CC=C(O)C=C3N=2)=N1 IGXWBGJHJZYPQS-SSDOTTSWSA-N 0.000 description 1
- QNAYBMKLOCPYGJ-UWTATZPHSA-N D-alanine Chemical compound C[C@@H](N)C(O)=O QNAYBMKLOCPYGJ-UWTATZPHSA-N 0.000 description 1
- QNAYBMKLOCPYGJ-UHFFFAOYSA-N D-alpha-Ala Natural products CC([NH3+])C([O-])=O QNAYBMKLOCPYGJ-UHFFFAOYSA-N 0.000 description 1
- 238000012270 DNA recombination Methods 0.000 description 1
- 108090000626 DNA-directed RNA polymerases Proteins 0.000 description 1
- 102000004163 DNA-directed RNA polymerases Human genes 0.000 description 1
- 108010092160 Dactinomycin Proteins 0.000 description 1
- XPDXVDYUQZHFPV-UHFFFAOYSA-N Dansyl Chloride Chemical compound C1=CC=C2C(N(C)C)=CC=CC2=C1S(Cl)(=O)=O XPDXVDYUQZHFPV-UHFFFAOYSA-N 0.000 description 1
- CYCGRDQQIOGCKX-UHFFFAOYSA-N Dehydro-luciferin Natural products OC(=O)C1=CSC(C=2SC3=CC(O)=CC=C3N=2)=N1 CYCGRDQQIOGCKX-UHFFFAOYSA-N 0.000 description 1
- 241001461743 Deltacoronavirus Species 0.000 description 1
- YZCKVEUIGOORGS-OUBTZVSYSA-N Deuterium Chemical group [2H] YZCKVEUIGOORGS-OUBTZVSYSA-N 0.000 description 1
- 229920002307 Dextran Polymers 0.000 description 1
- BWGNESOTFCXPMA-UHFFFAOYSA-N Dihydrogen disulfide Chemical compound SS BWGNESOTFCXPMA-UHFFFAOYSA-N 0.000 description 1
- 108060006698 EGF receptor Proteins 0.000 description 1
- 206010014561 Emphysema Diseases 0.000 description 1
- 108700041152 Endoplasmic Reticulum Chaperone BiP Proteins 0.000 description 1
- 102100021451 Endoplasmic reticulum chaperone BiP Human genes 0.000 description 1
- 101710204837 Envelope small membrane protein Proteins 0.000 description 1
- 102400001368 Epidermal growth factor Human genes 0.000 description 1
- 101800003838 Epidermal growth factor Proteins 0.000 description 1
- 241000701959 Escherichia virus Lambda Species 0.000 description 1
- FCEXWTOTHXCQCQ-UHFFFAOYSA-N Ethoxydihydrosanguinarine Natural products C12=CC=C3OCOC3=C2C(OCC)N(C)C(C2=C3)=C1C=CC2=CC1=C3OCO1 FCEXWTOTHXCQCQ-UHFFFAOYSA-N 0.000 description 1
- 241000711475 Feline infectious peritonitis virus Species 0.000 description 1
- 241000282326 Felis catus Species 0.000 description 1
- 102000008857 Ferritin Human genes 0.000 description 1
- 108050000784 Ferritin Proteins 0.000 description 1
- 238000008416 Ferritin Methods 0.000 description 1
- NJJDBBUWWOAOLD-UHFFFAOYSA-N Ferulenol Natural products C1=CC=C2OC(=O)C(CC=C(C)CCC=C(C)CCC=C(C)C)=C(O)C2=C1 NJJDBBUWWOAOLD-UHFFFAOYSA-N 0.000 description 1
- 102000009123 Fibrin Human genes 0.000 description 1
- 108010073385 Fibrin Proteins 0.000 description 1
- BWGVNKXGVNDBDI-UHFFFAOYSA-N Fibrin monomer Chemical compound CNC(=O)CNC(=O)CN BWGVNKXGVNDBDI-UHFFFAOYSA-N 0.000 description 1
- BJGNCJDXODQBOB-UHFFFAOYSA-N Fivefly Luciferin Natural products OC(=O)C1CSC(C=2SC3=CC(O)=CC=C3N=2)=N1 BJGNCJDXODQBOB-UHFFFAOYSA-N 0.000 description 1
- 241000700662 Fowlpox virus Species 0.000 description 1
- 241000233866 Fungi Species 0.000 description 1
- 241000287828 Gallus gallus Species 0.000 description 1
- 241000008920 Gammacoronavirus Species 0.000 description 1
- JRZJKWGQFNTSRN-UHFFFAOYSA-N Geldanamycin Natural products C1C(C)CC(OC)C(O)C(C)C=C(C)C(OC(N)=O)C(OC)CCC=C(C)C(=O)NC2=CC(=O)C(OC)=C1C2=O JRZJKWGQFNTSRN-UHFFFAOYSA-N 0.000 description 1
- 229930182566 Gentamicin Natural products 0.000 description 1
- CEAZRRDELHUEMR-URQXQFDESA-N Gentamicin Chemical compound O1[C@H](C(C)NC)CC[C@@H](N)[C@H]1O[C@H]1[C@H](O)[C@@H](O[C@@H]2[C@@H]([C@@H](NC)[C@@](C)(O)CO2)O)[C@H](N)C[C@@H]1N CEAZRRDELHUEMR-URQXQFDESA-N 0.000 description 1
- 229930195518 Glucopiericidin Natural products 0.000 description 1
- 108010026389 Gramicidin Proteins 0.000 description 1
- 108060003393 Granulin Proteins 0.000 description 1
- 102000004269 Granulocyte Colony-Stimulating Factor Human genes 0.000 description 1
- 108010017080 Granulocyte Colony-Stimulating Factor Proteins 0.000 description 1
- 108010017213 Granulocyte-Macrophage Colony-Stimulating Factor Proteins 0.000 description 1
- 102100039620 Granulocyte-macrophage colony-stimulating factor Human genes 0.000 description 1
- 206010053759 Growth retardation Diseases 0.000 description 1
- 239000007995 HEPES buffer Substances 0.000 description 1
- 229940122440 HIV protease inhibitor Drugs 0.000 description 1
- 108010072471 HTI-286 Proteins 0.000 description 1
- 206010019233 Headaches Diseases 0.000 description 1
- 208000002250 Hematologic Neoplasms Diseases 0.000 description 1
- 241000700721 Hepatitis B virus Species 0.000 description 1
- ISZXEMUWHQLLTC-UHFFFAOYSA-N Herboxidiene Natural products COC(C(C)O)C(C)C1OC1(C)CC(C)C=CC=C(C)C1C(C)CCC(CC(O)=O)O1 ISZXEMUWHQLLTC-UHFFFAOYSA-N 0.000 description 1
- 208000009889 Herpes Simplex Diseases 0.000 description 1
- 102100038030 High affinity immunoglobulin alpha and immunoglobulin mu Fc receptor Human genes 0.000 description 1
- 101710136666 High affinity immunoglobulin alpha and immunoglobulin mu Fc receptor Proteins 0.000 description 1
- 101000897480 Homo sapiens C-C motif chemokine 2 Proteins 0.000 description 1
- 101000611183 Homo sapiens Tumor necrosis factor Proteins 0.000 description 1
- 108010001336 Horseradish Peroxidase Proteins 0.000 description 1
- 241000701109 Human adenovirus 2 Species 0.000 description 1
- 241000419562 Human betacoronavirus 2c EMC/2012 Species 0.000 description 1
- 241000695864 Human betacoronavirus 2c Jordan-N3/2012 Species 0.000 description 1
- 241000701024 Human betaherpesvirus 5 Species 0.000 description 1
- UGIOXKVQFJIRMZ-UHFFFAOYSA-N Hygrolidin Natural products C1C(OC(=O)C=CC(O)=O)C(C)C(CC)OC1(O)C(C)C(O)C(C)C1C(OC)C=CC=C(C)CC(C)C(O)C(C)C=C(C)C=C(C)C(=O)O1 UGIOXKVQFJIRMZ-UHFFFAOYSA-N 0.000 description 1
- 201000009794 Idiopathic Pulmonary Fibrosis Diseases 0.000 description 1
- 108010079585 Immunoglobulin Subunits Proteins 0.000 description 1
- 102000012745 Immunoglobulin Subunits Human genes 0.000 description 1
- 102000003996 Interferon-beta Human genes 0.000 description 1
- 108090000467 Interferon-beta Proteins 0.000 description 1
- 108091092195 Intron Proteins 0.000 description 1
- OFFWOVJBSQMVPI-RMLGOCCBSA-N Kaletra Chemical compound N1([C@@H](C(C)C)C(=O)N[C@H](C[C@H](O)[C@H](CC=2C=CC=CC=2)NC(=O)COC=2C(=CC=CC=2C)C)CC=2C=CC=CC=2)CCCNC1=O.N([C@@H](C(C)C)C(=O)N[C@H](C[C@H](O)[C@H](CC=1C=CC=CC=1)NC(=O)OCC=1SC=NC=1)CC=1C=CC=CC=1)C(=O)N(C)CC1=CSC(C(C)C)=N1 OFFWOVJBSQMVPI-RMLGOCCBSA-N 0.000 description 1
- ZDXPYRJPNDTMRX-VKHMYHEASA-N L-glutamine Chemical compound OC(=O)[C@@H](N)CCC(N)=O ZDXPYRJPNDTMRX-VKHMYHEASA-N 0.000 description 1
- KZSNJWFQEVHDMF-BYPYZUCNSA-N L-valine Chemical compound CC(C)[C@H](N)C(O)=O KZSNJWFQEVHDMF-BYPYZUCNSA-N 0.000 description 1
- DAQAKHDKYAWHCG-UHFFFAOYSA-N Lactacystin Natural products CC(=O)NC(C(O)=O)CSC(=O)C1(C(O)C(C)C)NC(=O)C(C)C1O DAQAKHDKYAWHCG-UHFFFAOYSA-N 0.000 description 1
- JVTAAEKCZFNVCJ-UHFFFAOYSA-M Lactate Chemical compound CC(O)C([O-])=O JVTAAEKCZFNVCJ-UHFFFAOYSA-M 0.000 description 1
- 241000270322 Lepidosauria Species 0.000 description 1
- 108060001084 Luciferase Proteins 0.000 description 1
- 239000005089 Luciferase Substances 0.000 description 1
- DDWFXDSYGUXRAY-UHFFFAOYSA-N Luciferin Natural products CCc1c(C)c(CC2NC(=O)C(=C2C=C)C)[nH]c1Cc3[nH]c4C(=C5/NC(CC(=O)O)C(C)C5CC(=O)O)CC(=O)c4c3C DDWFXDSYGUXRAY-UHFFFAOYSA-N 0.000 description 1
- 206010025323 Lymphomas Diseases 0.000 description 1
- 101710145006 Lysis protein Proteins 0.000 description 1
- FYYHWMGAXLPEAU-UHFFFAOYSA-N Magnesium Chemical compound [Mg] FYYHWMGAXLPEAU-UHFFFAOYSA-N 0.000 description 1
- 241000124008 Mammalia Species 0.000 description 1
- 101710085938 Matrix protein Proteins 0.000 description 1
- 102000012750 Membrane Glycoproteins Human genes 0.000 description 1
- 108010090054 Membrane Glycoproteins Proteins 0.000 description 1
- 101710127721 Membrane protein Proteins 0.000 description 1
- MNASOWORUXKEPP-UHFFFAOYSA-N Microcolin B Natural products CCCCC(C)CC(C)C(=O)N(C)C(CC(C)C)C(=O)NC(C(C)OC(C)=O)C(=O)N(C)C(C(C)C)C(=O)N1CCCC1C(=O)N1C(=O)C=CC1C MNASOWORUXKEPP-UHFFFAOYSA-N 0.000 description 1
- 206010028124 Mucosal ulceration Diseases 0.000 description 1
- 241001529936 Murinae Species 0.000 description 1
- 208000000112 Myalgia Diseases 0.000 description 1
- 241000713883 Myeloproliferative sarcoma virus Species 0.000 description 1
- ZJZWZIXSGNFWQQ-UHFFFAOYSA-N N-[4-[[[4-(4-methoxyphenyl)-4-oxanyl]methylamino]-oxomethyl]phenyl]-2-furancarboxamide Chemical compound C1=CC(OC)=CC=C1C1(CNC(=O)C=2C=CC(NC(=O)C=3OC=CC=3)=CC=2)CCOCC1 ZJZWZIXSGNFWQQ-UHFFFAOYSA-N 0.000 description 1
- 125000000729 N-terminal amino-acid group Chemical group 0.000 description 1
- 108091007491 NSP3 Papain-like protease domains Proteins 0.000 description 1
- 206010028851 Necrosis Diseases 0.000 description 1
- 241000918515 Neoromicia Species 0.000 description 1
- 239000000020 Nitrocellulose Substances 0.000 description 1
- 101710141454 Nucleoprotein Proteins 0.000 description 1
- VYVUCONKFCPKEV-UHFFFAOYSA-N Okilactomycin Natural products CC1CCCC2C=C(C(C)CC23OC(=O)C4(C)C3CC(C(C)C1)C(=O)C4=O)C(=O)O VYVUCONKFCPKEV-UHFFFAOYSA-N 0.000 description 1
- 108020005187 Oligonucleotide Probes Proteins 0.000 description 1
- 108010038807 Oligopeptides Proteins 0.000 description 1
- 102000015636 Oligopeptides Human genes 0.000 description 1
- 102100021079 Ornithine decarboxylase Human genes 0.000 description 1
- 108700005126 Ornithine decarboxylases Proteins 0.000 description 1
- 206010068319 Oropharyngeal pain Diseases 0.000 description 1
- 206010033307 Overweight Diseases 0.000 description 1
- 238000012408 PCR amplification Methods 0.000 description 1
- 229910019142 PO4 Inorganic materials 0.000 description 1
- 102000016387 Pancreatic elastase Human genes 0.000 description 1
- 108010067372 Pancreatic elastase Proteins 0.000 description 1
- 241000282320 Panthera leo Species 0.000 description 1
- 241000282376 Panthera tigris Species 0.000 description 1
- 241001494479 Pecora Species 0.000 description 1
- 108090000284 Pepsin A Proteins 0.000 description 1
- 102000057297 Pepsin A Human genes 0.000 description 1
- 241000009328 Perro Species 0.000 description 1
- 101800000891 Phallacidin Proteins 0.000 description 1
- 108010009711 Phalloidine Proteins 0.000 description 1
- 201000007100 Pharyngitis Diseases 0.000 description 1
- OAICVXFJPJFONN-OUBTZVSYSA-N Phosphorus-32 Chemical compound [32P] OAICVXFJPJFONN-OUBTZVSYSA-N 0.000 description 1
- 108010004729 Phycoerythrin Proteins 0.000 description 1
- 229930182661 Piericidin Natural products 0.000 description 1
- 241001125039 Pipistrellus Species 0.000 description 1
- 241000008909 Pipistrellus bat coronavirus HKU5 Species 0.000 description 1
- OWRCRVABZYJUES-UHFFFAOYSA-N Polyketomycin Natural products COC1=CC(=O)c2c(O)c3C(=O)C4(O)C(=O)C(=C(/C)O)C(=O)C(OC5CCC(OC6CC(C)(O)C(OC(=O)c7c(C)ccc(C)c7O)C(C)O6)C(C)O5)C4(O)Cc3c(C)c2C1=O OWRCRVABZYJUES-UHFFFAOYSA-N 0.000 description 1
- 241001505332 Polyomavirus sp. Species 0.000 description 1
- 208000020584 Polyploidy Diseases 0.000 description 1
- 239000004372 Polyvinyl alcohol Substances 0.000 description 1
- 241001135549 Porcine epidemic diarrhea virus Species 0.000 description 1
- 241000156302 Porcine hemagglutinating encephalomyelitis virus Species 0.000 description 1
- 241001135989 Porcine reproductive and respiratory syndrome virus Species 0.000 description 1
- 208000031951 Primary immunodeficiency Diseases 0.000 description 1
- 102000006010 Protein Disulfide-Isomerase Human genes 0.000 description 1
- 108010029485 Protein Isoforms Proteins 0.000 description 1
- 102000001708 Protein Isoforms Human genes 0.000 description 1
- 101000933967 Pseudomonas phage KPP25 Major capsid protein Proteins 0.000 description 1
- GZAYJWNDYQINEI-OBSDVKHNSA-N Pseurotin D Natural products CC[C@@H](O)C=C[C@H](O)C1=C(C)C(=O)[C@@]2(O1)[C@@H](O)[C@](NC2=O)(OC)C(=O)c3ccccc3 GZAYJWNDYQINEI-OBSDVKHNSA-N 0.000 description 1
- 229930188256 Puwainaphycin Natural products 0.000 description 1
- KIPSHYPQCJYONU-UHFFFAOYSA-N Quinaldopeptin Natural products CC1NC(=O)C2CCCCN2C(=O)CN(C)C(=O)CNC(=O)C2CCCCN2C(=O)C(NC(=O)C=2C(=CC3=CC=CC=C3N=2)O)C(C)NC(=O)C2CCCCN2C(=O)CN(C)C(=O)CNC(=O)C2CCCCN2C(=O)C1NC(=O)C1=NC2=CC=CC=C2C=C1O KIPSHYPQCJYONU-UHFFFAOYSA-N 0.000 description 1
- 102000004879 Racemases and epimerases Human genes 0.000 description 1
- 108090001066 Racemases and epimerases Proteins 0.000 description 1
- QEHOIJJIZXRMAN-UHFFFAOYSA-N Rebeccamycin Natural products OC1C(O)C(OC)C(CO)OC1N1C2=C3NC4=C(Cl)C=CC=C4C3=C3C(=O)NC(=O)C3=C2C2=CC=CC(Cl)=C21 QEHOIJJIZXRMAN-UHFFFAOYSA-N 0.000 description 1
- XWNMORIHKRROGW-UHFFFAOYSA-N Ro 5-3335 Chemical compound C12=CC(Cl)=CC=C2NC(=O)CN=C1C1=CC=CN1 XWNMORIHKRROGW-UHFFFAOYSA-N 0.000 description 1
- 241000714474 Rous sarcoma virus Species 0.000 description 1
- 208000037847 SARS-CoV-2-infection Diseases 0.000 description 1
- 229930183496 Safracin Natural products 0.000 description 1
- 241000700584 Simplexvirus Species 0.000 description 1
- 101710147732 Small envelope protein Proteins 0.000 description 1
- 108091027967 Small hairpin RNA Proteins 0.000 description 1
- 108020004459 Small interfering RNA Proteins 0.000 description 1
- 108091081024 Start codon Proteins 0.000 description 1
- 108091027544 Subgenomic mRNA Proteins 0.000 description 1
- NINIDFKCEFEMDL-AKLPVKDBSA-N Sulfur-35 Chemical compound [35S] NINIDFKCEFEMDL-AKLPVKDBSA-N 0.000 description 1
- 241000282898 Sus scrofa Species 0.000 description 1
- 108091033399 Telomestatin Proteins 0.000 description 1
- GUGOEEXESWIERI-UHFFFAOYSA-N Terfenadine Chemical compound C1=CC(C(C)(C)C)=CC=C1C(O)CCCN1CCC(C(O)(C=2C=CC=CC=2)C=2C=CC=CC=2)CC1 GUGOEEXESWIERI-UHFFFAOYSA-N 0.000 description 1
- 108020005038 Terminator Codon Proteins 0.000 description 1
- 239000004098 Tetracycline Substances 0.000 description 1
- 208000035199 Tetraploidy Diseases 0.000 description 1
- FTGOWEQDZZMPNJ-GUMZKTDUSA-N Tolytoxin Chemical compound C([C@@]12[C@@H](OC)C[C@H]3O[C@@H](C=CC3)C[C@H]([C@@H](O)/C=C(\C)/C=C/C(=O)O[C@@H]([C@H]([C@H](OC)C[C@@H]2OC)C)[C@@H](C)[C@@H](O)[C@@H](C)CCC(=O)[C@H](C)[C@@H]([C@H](C)\C=C\N(C)C=O)OC)OC)O1 FTGOWEQDZZMPNJ-GUMZKTDUSA-N 0.000 description 1
- 102000040945 Transcription factor Human genes 0.000 description 1
- 108091023040 Transcription factor Proteins 0.000 description 1
- 108700029229 Transcriptional Regulatory Elements Proteins 0.000 description 1
- DFBIRQPKNDILPW-CIVMWXNOSA-N Triptolide Chemical compound O=C1OCC([C@@H]2C3)=C1CC[C@]2(C)[C@]12O[C@H]1[C@@H]1O[C@]1(C(C)C)[C@@H](O)[C@]21[C@H]3O1 DFBIRQPKNDILPW-CIVMWXNOSA-N 0.000 description 1
- YZCKVEUIGOORGS-NJFSPNSNSA-N Tritium Chemical compound [3H] YZCKVEUIGOORGS-NJFSPNSNSA-N 0.000 description 1
- 102100040247 Tumor necrosis factor Human genes 0.000 description 1
- 241000008908 Tylonycteris bat coronavirus HKU4 Species 0.000 description 1
- 206010067584 Type 1 diabetes mellitus Diseases 0.000 description 1
- 208000025865 Ulcer Diseases 0.000 description 1
- KZSNJWFQEVHDMF-UHFFFAOYSA-N Valine Natural products CC(C)C(N)C(O)=O KZSNJWFQEVHDMF-UHFFFAOYSA-N 0.000 description 1
- 229940123627 Viral replication inhibitor Drugs 0.000 description 1
- 108010084455 Zeocin Proteins 0.000 description 1
- PVNFMCBFDPTNQI-UIBOPQHZSA-N [(1S,2R,5S,6S,16E,18E,20R,21S)-11-chloro-21-hydroxy-12,20-dimethoxy-2,5,9,16-tetramethyl-8,23-dioxo-4,24-dioxa-9,22-diazatetracyclo[19.3.1.110,14.03,5]hexacosa-10,12,14(26),16,18-pentaen-6-yl] acetate [(1S,2R,5S,6S,16E,18E,20R,21S)-11-chloro-21-hydroxy-12,20-dimethoxy-2,5,9,16-tetramethyl-8,23-dioxo-4,24-dioxa-9,22-diazatetracyclo[19.3.1.110,14.03,5]hexacosa-10,12,14(26),16,18-pentaen-6-yl] 3-methylbutanoate [(1S,2R,5S,6S,16E,18E,20R,21S)-11-chloro-21-hydroxy-12,20-dimethoxy-2,5,9,16-tetramethyl-8,23-dioxo-4,24-dioxa-9,22-diazatetracyclo[19.3.1.110,14.03,5]hexacosa-10,12,14(26),16,18-pentaen-6-yl] 2-methylpropanoate [(1S,2R,5S,6S,16E,18E,20R,21S)-11-chloro-21-hydroxy-12,20-dimethoxy-2,5,9,16-tetramethyl-8,23-dioxo-4,24-dioxa-9,22-diazatetracyclo[19.3.1.110,14.03,5]hexacosa-10,12,14(26),16,18-pentaen-6-yl] propanoate Chemical compound CO[C@@H]1\C=C\C=C(C)\Cc2cc(OC)c(Cl)c(c2)N(C)C(=O)C[C@H](OC(C)=O)[C@]2(C)OC2[C@H](C)[C@@H]2C[C@@]1(O)NC(=O)O2.CCC(=O)O[C@H]1CC(=O)N(C)c2cc(C\C(C)=C\C=C\[C@@H](OC)[C@@]3(O)C[C@H](OC(=O)N3)[C@@H](C)C3O[C@@]13C)cc(OC)c2Cl.CO[C@@H]1\C=C\C=C(C)\Cc2cc(OC)c(Cl)c(c2)N(C)C(=O)C[C@H](OC(=O)C(C)C)[C@]2(C)OC2[C@H](C)[C@@H]2C[C@@]1(O)NC(=O)O2.CO[C@@H]1\C=C\C=C(C)\Cc2cc(OC)c(Cl)c(c2)N(C)C(=O)C[C@H](OC(=O)CC(C)C)[C@]2(C)OC2[C@H](C)[C@@H]2C[C@@]1(O)NC(=O)O2 PVNFMCBFDPTNQI-UIBOPQHZSA-N 0.000 description 1
- MNASOWORUXKEPP-DEZPKOTESA-N [(2s,3s)-3-[[(2s)-2-[[(2r,4r)-2,4-dimethyloctanoyl]-methylamino]-4-methylpentanoyl]amino]-4-[methyl-[(2s)-3-methyl-1-[(2s)-2-[(2s)-2-methyl-5-oxo-2h-pyrrole-1-carbonyl]pyrrolidin-1-yl]-1-oxobutan-2-yl]amino]-4-oxobutan-2-yl] acetate Chemical compound CCCC[C@@H](C)C[C@@H](C)C(=O)N(C)[C@@H](CC(C)C)C(=O)N[C@@H]([C@H](C)OC(C)=O)C(=O)N(C)[C@@H](C(C)C)C(=O)N1CCC[C@H]1C(=O)N1C(=O)C=C[C@@H]1C MNASOWORUXKEPP-DEZPKOTESA-N 0.000 description 1
- UNPSMUMJWYZQHC-CYVBNFEDSA-N [(3r,5s,6r,7s,8e,10s,11s,12z,14e)-21-[3-(dimethylamino)propylamino]-6-hydroxy-5,11-dimethoxy-3,7,9,15-tetramethyl-16,20,22-trioxo-17-azabicyclo[16.3.1]docosa-1(21),8,12,14,18-pentaen-10-yl] carbamate Chemical compound N1C(=O)\C(C)=C\C=C/[C@H](OC)[C@@H](OC(N)=O)\C(C)=C\[C@H](C)[C@@H](O)[C@@H](OC)C[C@H](C)CC2=C(NCCCN(C)C)C(=O)C=C1C2=O UNPSMUMJWYZQHC-CYVBNFEDSA-N 0.000 description 1
- 230000005856 abnormality Effects 0.000 description 1
- 229940022698 acetylcholinesterase Drugs 0.000 description 1
- 108020002494 acetyltransferase Proteins 0.000 description 1
- 102000005421 acetyltransferase Human genes 0.000 description 1
- 239000002253 acid Substances 0.000 description 1
- 229930183665 actinomycin Natural products 0.000 description 1
- 230000003213 activating effect Effects 0.000 description 1
- 239000012190 activator Substances 0.000 description 1
- 229960005305 adenosine Drugs 0.000 description 1
- 230000001464 adherent effect Effects 0.000 description 1
- 238000001261 affinity purification Methods 0.000 description 1
- QZBCDLLHQSDFIG-ZEVBXJOLSA-N agrochelin Chemical compound CCCCCC1=CC=CC(O)=C1C1=N[C@@H]([C@H]2N([C@H]([C@@H](O)C(C)(C)C(O)=O)CS2)C)CS1 QZBCDLLHQSDFIG-ZEVBXJOLSA-N 0.000 description 1
- QZBCDLLHQSDFIG-UHFFFAOYSA-N agrochelin Natural products CCCCCc1cccc(O)c1C1=NC(CS1)C1SCC(C(O)C(C)(C)C(O)=O)N1C QZBCDLLHQSDFIG-UHFFFAOYSA-N 0.000 description 1
- SSNQAUBBJYCSMY-UHFFFAOYSA-N aigialomycin A Natural products C12OC2CC(O)C(O)C(=O)C=CCC(C)OC(=O)C=2C1=CC(OC)=CC=2O SSNQAUBBJYCSMY-UHFFFAOYSA-N 0.000 description 1
- 229940100198 alkylating agent Drugs 0.000 description 1
- 239000002168 alkylating agent Substances 0.000 description 1
- 108010026331 alpha-Fetoproteins Proteins 0.000 description 1
- CIORWBWIBBPXCG-JZTFPUPKSA-N amanitin Chemical compound O=C1N[C@@H](CC(N)=O)C(=O)N2CC(O)C[C@H]2C(=O)N[C@@H](C(C)[C@@H](O)CO)C(=O)N[C@@H](C2)C(=O)NCC(=O)N[C@@H](C(C)CC)C(=O)NCC(=O)N[C@H]1CS(=O)C1=C2C2=CC=C(O)C=C2N1 CIORWBWIBBPXCG-JZTFPUPKSA-N 0.000 description 1
- 125000003368 amide group Chemical group 0.000 description 1
- 150000001408 amides Chemical group 0.000 description 1
- 150000001412 amines Chemical class 0.000 description 1
- 210000004102 animal cell Anatomy 0.000 description 1
- 238000005571 anion exchange chromatography Methods 0.000 description 1
- 230000000507 anthelmentic effect Effects 0.000 description 1
- 230000002391 anti-complement effect Effects 0.000 description 1
- 230000003510 anti-fibrotic effect Effects 0.000 description 1
- 230000001387 anti-histamine Effects 0.000 description 1
- 230000003110 anti-inflammatory effect Effects 0.000 description 1
- 230000000340 anti-metabolite Effects 0.000 description 1
- 230000009830 antibody antigen interaction Effects 0.000 description 1
- 230000005875 antibody response Effects 0.000 description 1
- 108010008730 anticomplement Proteins 0.000 description 1
- 229940100197 antimetabolite Drugs 0.000 description 1
- 239000002256 antimetabolite Substances 0.000 description 1
- 239000004599 antimicrobial Substances 0.000 description 1
- 229940121357 antivirals Drugs 0.000 description 1
- 238000002617 apheresis Methods 0.000 description 1
- NOFOAYPPHIUXJR-APNQCZIXSA-N aphidicolin Chemical compound C1[C@@]23[C@@]4(C)CC[C@@H](O)[C@@](C)(CO)[C@@H]4CC[C@H]3C[C@H]1[C@](CO)(O)CC2 NOFOAYPPHIUXJR-APNQCZIXSA-N 0.000 description 1
- SEKZNWAQALMJNH-YZUCACDQSA-N aphidicolin Natural products C[C@]1(CO)CC[C@]23C[C@H]1C[C@@H]2CC[C@H]4[C@](C)(CO)[C@H](O)CC[C@]34C SEKZNWAQALMJNH-YZUCACDQSA-N 0.000 description 1
- 230000001640 apoptogenic effect Effects 0.000 description 1
- WILMROCKORZEMQ-UHFFFAOYSA-N apoptolidin Natural products C1C(C)(O)C(OC2OC(C)C(O)C(OC)C2)C(C)OC1OC(COC)CC(C(C(O)C1C)C)OC1(O)C(O)C(OC(=O)C(C)=CC(C)=CC(C)=CC1C)CC(OC)C(O)CCC=C(C)C=CC1OC1OC(C)C(OC)C(O)C1O WILMROCKORZEMQ-UHFFFAOYSA-N 0.000 description 1
- 238000013459 approach Methods 0.000 description 1
- 108010044540 auristatin Proteins 0.000 description 1
- 229960000190 bacillus calmette–guérin vaccine Drugs 0.000 description 1
- 229930192649 bafilomycin Natural products 0.000 description 1
- XDHNQDDQEHDUTM-UHFFFAOYSA-N bafliomycin A1 Natural products COC1C=CC=C(C)CC(C)C(O)C(C)C=C(C)C=C(OC)C(=O)OC1C(C)C(O)C(C)C1(O)OC(C(C)C)C(C)C(O)C1 XDHNQDDQEHDUTM-UHFFFAOYSA-N 0.000 description 1
- 238000010009 beating Methods 0.000 description 1
- 208000013404 behavioral symptom Diseases 0.000 description 1
- 239000002876 beta blocker Substances 0.000 description 1
- 229940097320 beta blocking agent Drugs 0.000 description 1
- IQFYYKKMVGJFEH-UHFFFAOYSA-N beta-L-thymidine Natural products O=C1NC(=O)C(C)=CN1C1OC(CO)C(O)C1 IQFYYKKMVGJFEH-UHFFFAOYSA-N 0.000 description 1
- 239000012472 biological sample Substances 0.000 description 1
- 230000017531 blood circulation Effects 0.000 description 1
- 210000001124 body fluid Anatomy 0.000 description 1
- 238000006664 bond formation reaction Methods 0.000 description 1
- 210000001185 bone marrow Anatomy 0.000 description 1
- 108010006025 bovine growth hormone Proteins 0.000 description 1
- 229940098773 bovine serum albumin Drugs 0.000 description 1
- 229940124630 bronchodilator Drugs 0.000 description 1
- 239000000872 buffer Substances 0.000 description 1
- 239000011575 calcium Substances 0.000 description 1
- 229910052791 calcium Inorganic materials 0.000 description 1
- 229930195731 calicheamicin Natural products 0.000 description 1
- HXCHCVDVKSCDHU-LULTVBGHSA-N calicheamicin Chemical compound C1[C@H](OC)[C@@H](NCC)CO[C@H]1O[C@H]1[C@H](O[C@@H]2C\3=C(NC(=O)OC)C(=O)C[C@](C/3=C/CSSSC)(O)C#C\C=C/C#C2)O[C@H](C)[C@@H](NO[C@@H]2O[C@H](C)[C@@H](SC(=O)C=3C(=C(OC)C(O[C@H]4[C@@H]([C@H](OC)[C@@H](O)[C@H](C)O4)O)=C(I)C=3C)OC)[C@@H](O)C2)[C@@H]1O HXCHCVDVKSCDHU-LULTVBGHSA-N 0.000 description 1
- 201000011510 cancer Diseases 0.000 description 1
- 210000000234 capsid Anatomy 0.000 description 1
- 125000000837 carbohydrate group Chemical group 0.000 description 1
- 239000001768 carboxy methyl cellulose Substances 0.000 description 1
- 235000010948 carboxy methyl cellulose Nutrition 0.000 description 1
- 239000008112 carboxymethyl-cellulose Substances 0.000 description 1
- 230000015556 catabolic process Effects 0.000 description 1
- 239000003054 catalyst Substances 0.000 description 1
- 230000003197 catalytic effect Effects 0.000 description 1
- 238000005277 cation exchange chromatography Methods 0.000 description 1
- 125000002091 cationic group Chemical group 0.000 description 1
- 238000004113 cell culture Methods 0.000 description 1
- 210000000170 cell membrane Anatomy 0.000 description 1
- 230000036755 cellular response Effects 0.000 description 1
- 238000005119 centrifugation Methods 0.000 description 1
- 239000000919 ceramic Substances 0.000 description 1
- 206010008129 cerebral palsy Diseases 0.000 description 1
- PZPPOCZWRGNKIR-PNVYSBBASA-N chaetocin Chemical compound N([C@@H]1N2C(=O)[C@]3(CO)SS[C@]2(C(N3C)=O)C2)C3=CC=CC=C3[C@]12[C@@]12C[C@]3(SS4)C(=O)N(C)[C@]4(CO)C(=O)N3[C@H]2NC2=CC=CC=C12 PZPPOCZWRGNKIR-PNVYSBBASA-N 0.000 description 1
- PZPPOCZWRGNKIR-UHFFFAOYSA-N chaetocin Natural products C1C2(C(N3C)=O)SSC3(CO)C(=O)N2C2NC3=CC=CC=C3C21C12CC3(SS4)C(=O)N(C)C4(CO)C(=O)N3C2NC2=CC=CC=C12 PZPPOCZWRGNKIR-UHFFFAOYSA-N 0.000 description 1
- 229930191237 chaetoglobosin Natural products 0.000 description 1
- PRSOPHSUKXJKIM-CFBAGHHKSA-N chembl174839 Chemical compound C1=CC=C2C(=O)C(C/C=C(C)/CC/C=C(C)/CCC=C(C)C)=C(O)OC2=C1 PRSOPHSUKXJKIM-CFBAGHHKSA-N 0.000 description 1
- KIPSHYPQCJYONU-GVPFWXHKSA-N chembl1994663 Chemical compound N([C@@H](C(C(=O)N1CCCC[C@H]1C(=O)NCC(=O)N(C)CC(=O)N1CCCCC1C(=O)N[C@@H]1C)NC(=O)C=2C(=CC3=CC=CC=C3N=2)O)C)C(=O)C2CCCCN2C(=O)CN(C)C(=O)CNC(=O)C2CCCCN2C(=O)[C@H]1NC(=O)C1=NC2=CC=CC=C2C=C1O KIPSHYPQCJYONU-GVPFWXHKSA-N 0.000 description 1
- 238000007385 chemical modification Methods 0.000 description 1
- 238000006243 chemical reaction Methods 0.000 description 1
- 239000003153 chemical reaction reagent Substances 0.000 description 1
- 239000003638 chemical reducing agent Substances 0.000 description 1
- 238000002512 chemotherapy Methods 0.000 description 1
- 108700023145 chlamydocin Proteins 0.000 description 1
- 239000013611 chromosomal DNA Substances 0.000 description 1
- 208000007451 chronic bronchitis Diseases 0.000 description 1
- 229960002376 chymotrypsin Drugs 0.000 description 1
- 230000004087 circulation Effects 0.000 description 1
- 230000004186 co-expression Effects 0.000 description 1
- 229960001338 colchicine Drugs 0.000 description 1
- 238000004440 column chromatography Methods 0.000 description 1
- LGZKGOGODCLQHG-UHFFFAOYSA-N combretastatin Natural products C1=C(O)C(OC)=CC=C1CC(O)C1=CC(OC)=C(OC)C(OC)=C1 LGZKGOGODCLQHG-UHFFFAOYSA-N 0.000 description 1
- 230000002860 competitive effect Effects 0.000 description 1
- 208000028831 congenital heart disease Diseases 0.000 description 1
- 230000001268 conjugating effect Effects 0.000 description 1
- 238000010276 construction Methods 0.000 description 1
- 238000011109 contamination Methods 0.000 description 1
- 238000007796 conventional method Methods 0.000 description 1
- 229920001577 copolymer Polymers 0.000 description 1
- OFEZSBMBBKLLBJ-BAJZRUMYSA-N cordycepin Chemical compound C1=NC=2C(N)=NC=NC=2N1[C@@H]1O[C@H](CO)C[C@H]1O OFEZSBMBBKLLBJ-BAJZRUMYSA-N 0.000 description 1
- OFEZSBMBBKLLBJ-UHFFFAOYSA-N cordycepine Natural products C1=NC=2C(N)=NC=NC=2N1C1OC(CO)CC1O OFEZSBMBBKLLBJ-UHFFFAOYSA-N 0.000 description 1
- 208000029078 coronary artery disease Diseases 0.000 description 1
- 230000008878 coupling Effects 0.000 description 1
- 238000010168 coupling process Methods 0.000 description 1
- 238000005859 coupling reaction Methods 0.000 description 1
- 230000009260 cross reactivity Effects 0.000 description 1
- 230000005574 cross-species transmission Effects 0.000 description 1
- 150000001904 cucurbitacins Chemical class 0.000 description 1
- QASFUMOKHFSJGL-UHFFFAOYSA-N cyclopamine Natural products C1C=C2CC(O)CCC2(C)C(CC2=C3C)C1C2CCC13OC2CC(C)CNC2C1C QASFUMOKHFSJGL-UHFFFAOYSA-N 0.000 description 1
- 102000003675 cytokine receptors Human genes 0.000 description 1
- 108010057085 cytokine receptors Proteins 0.000 description 1
- 210000000172 cytosol Anatomy 0.000 description 1
- 230000001086 cytosolic effect Effects 0.000 description 1
- 239000002619 cytotoxin Substances 0.000 description 1
- STQGQHZAVUOBTE-VGBVRHCVSA-N daunorubicin Chemical compound O([C@H]1C[C@@](O)(CC=2C(O)=C3C(=O)C=4C=CC=C(C=4C(=O)C3=C(O)C=21)OC)C(C)=O)[C@H]1C[C@H](N)[C@H](O)[C@H](C)O1 STQGQHZAVUOBTE-VGBVRHCVSA-N 0.000 description 1
- 229960000975 daunorubicin Drugs 0.000 description 1
- 230000002950 deficient Effects 0.000 description 1
- 238000006731 degradation reaction Methods 0.000 description 1
- 230000001934 delay Effects 0.000 description 1
- 210000004443 dendritic cell Anatomy 0.000 description 1
- 230000001419 dependent effect Effects 0.000 description 1
- 238000011118 depth filtration Methods 0.000 description 1
- 238000001212 derivatisation Methods 0.000 description 1
- 238000013461 design Methods 0.000 description 1
- 235000005911 diet Nutrition 0.000 description 1
- 230000037213 diet Effects 0.000 description 1
- 238000001085 differential centrifugation Methods 0.000 description 1
- 238000002050 diffraction method Methods 0.000 description 1
- PIGAXYFCLPQWOD-UHFFFAOYSA-N dihydrocucurbitacin I Natural products CC12C(=O)CC3(C)C(C(C)(O)C(=O)CCC(C)(O)C)C(O)CC3(C)C1CC=C1C2C=C(O)C(=O)C1(C)C PIGAXYFCLPQWOD-UHFFFAOYSA-N 0.000 description 1
- AMRJKAQTDDKMCE-UHFFFAOYSA-N dolastatin Chemical compound CC(C)C(N(C)C)C(=O)NC(C(C)C)C(=O)N(C)C(C(C)C)C(OC)CC(=O)N1CCCC1C(OC)C(C)C(=O)NC(C=1SC=CN=1)CC1=CC=CC=C1 AMRJKAQTDDKMCE-UHFFFAOYSA-N 0.000 description 1
- 229930188854 dolastatin Natural products 0.000 description 1
- 230000003828 downregulation Effects 0.000 description 1
- 229960004679 doxorubicin Drugs 0.000 description 1
- 208000017574 dry cough Diseases 0.000 description 1
- 229960005501 duocarmycin Drugs 0.000 description 1
- VQNATVDKACXKTF-XELLLNAOSA-N duocarmycin Chemical compound COC1=C(OC)C(OC)=C2NC(C(=O)N3C4=CC(=O)C5=C([C@@]64C[C@@H]6C3)C=C(N5)C(=O)OC)=CC2=C1 VQNATVDKACXKTF-XELLLNAOSA-N 0.000 description 1
- 229930184221 duocarmycin Natural products 0.000 description 1
- 238000001493 electron microscopy Methods 0.000 description 1
- 238000002330 electrospray ionisation mass spectrometry Methods 0.000 description 1
- 238000010828 elution Methods 0.000 description 1
- 238000013504 emergency use authorization Methods 0.000 description 1
- 208000028208 end stage renal disease Diseases 0.000 description 1
- 201000000523 end stage renal failure Diseases 0.000 description 1
- 229930185143 englerin Natural products 0.000 description 1
- 230000002255 enzymatic effect Effects 0.000 description 1
- 229940116977 epidermal growth factor Drugs 0.000 description 1
- 230000001667 episodic effect Effects 0.000 description 1
- 229930013356 epothilone Natural products 0.000 description 1
- HESCAJZNRMSMJG-HGYUPSKWSA-N epothilone A Natural products O=C1[C@H](C)[C@H](O)[C@H](C)CCC[C@H]2O[C@H]2C[C@@H](/C(=C\c2nc(C)sc2)/C)OC(=O)C[C@H](O)C1(C)C HESCAJZNRMSMJG-HGYUPSKWSA-N 0.000 description 1
- HESCAJZNRMSMJG-KKQRBIROSA-N epothilone A Chemical compound C/C([C@@H]1C[C@@H]2O[C@@H]2CCC[C@@H]([C@@H]([C@@H](C)C(=O)C(C)(C)[C@@H](O)CC(=O)O1)O)C)=C\C1=CSC(C)=N1 HESCAJZNRMSMJG-KKQRBIROSA-N 0.000 description 1
- 150000003883 epothilone derivatives Chemical class 0.000 description 1
- 108010014507 erythroagglutinating phytohemagglutinin Proteins 0.000 description 1
- 239000003797 essential amino acid Substances 0.000 description 1
- 235000020776 essential amino acid Nutrition 0.000 description 1
- 238000011156 evaluation Methods 0.000 description 1
- 230000007717 exclusion Effects 0.000 description 1
- 206010016256 fatigue Diseases 0.000 description 1
- 210000003608 fece Anatomy 0.000 description 1
- 210000003754 fetus Anatomy 0.000 description 1
- 229950003499 fibrin Drugs 0.000 description 1
- 230000003176 fibrotic effect Effects 0.000 description 1
- 238000001914 filtration Methods 0.000 description 1
- 239000013020 final formulation Substances 0.000 description 1
- 238000005189 flocculation Methods 0.000 description 1
- 230000016615 flocculation Effects 0.000 description 1
- GNBHRKFJIUUOQI-UHFFFAOYSA-N fluorescein Chemical compound O1C(=O)C2=CC=CC=C2C21C1=CC=C(O)C=C1OC1=CC(O)=CC=C21 GNBHRKFJIUUOQI-UHFFFAOYSA-N 0.000 description 1
- MHMNJMPURVTYEJ-UHFFFAOYSA-N fluorescein-5-isothiocyanate Chemical compound O1C(=O)C2=CC(N=C=S)=CC=C2C21C1=CC=C(O)C=C1OC1=CC(O)=CC=C21 MHMNJMPURVTYEJ-UHFFFAOYSA-N 0.000 description 1
- 102000035175 foldases Human genes 0.000 description 1
- 108091005749 foldases Proteins 0.000 description 1
- 229960000936 fumagillin Drugs 0.000 description 1
- NGGMYCMLYOUNGM-CSDLUJIJSA-N fumagillin Chemical compound C([C@H]([C@H]([C@@H]1[C@]2(C)[C@H](O2)CC=C(C)C)OC)OC(=O)\C=C\C=C\C=C\C=C\C(O)=O)C[C@@]21CO2 NGGMYCMLYOUNGM-CSDLUJIJSA-N 0.000 description 1
- 125000000524 functional group Chemical group 0.000 description 1
- 108020001507 fusion proteins Proteins 0.000 description 1
- 102000037865 fusion proteins Human genes 0.000 description 1
- QTQAWLPCGQOSGP-GBTDJJJQSA-N geldanamycin Chemical compound N1C(=O)\C(C)=C/C=C\[C@@H](OC)[C@H](OC(N)=O)\C(C)=C/[C@@H](C)[C@@H](O)[C@H](OC)C[C@@H](C)CC2=C(OC)C(=O)C=C1C2=O QTQAWLPCGQOSGP-GBTDJJJQSA-N 0.000 description 1
- 238000010353 genetic engineering Methods 0.000 description 1
- 108060003196 globin Proteins 0.000 description 1
- 102000018146 globin Human genes 0.000 description 1
- 229930195712 glutamate Natural products 0.000 description 1
- ZDXPYRJPNDTMRX-UHFFFAOYSA-N glutamine Natural products OC(=O)C(N)CCC(N)=O ZDXPYRJPNDTMRX-UHFFFAOYSA-N 0.000 description 1
- 150000004676 glycans Chemical class 0.000 description 1
- PCHJSUWPFVWCPO-UHFFFAOYSA-N gold Chemical compound [Au] PCHJSUWPFVWCPO-UHFFFAOYSA-N 0.000 description 1
- 239000010931 gold Substances 0.000 description 1
- 229910052737 gold Inorganic materials 0.000 description 1
- 230000005484 gravity Effects 0.000 description 1
- 239000003102 growth factor Substances 0.000 description 1
- 238000001631 haemodialysis Methods 0.000 description 1
- 238000003306 harvesting Methods 0.000 description 1
- 210000003128 head Anatomy 0.000 description 1
- 231100000869 headache Toxicity 0.000 description 1
- 230000000322 hemodialysis Effects 0.000 description 1
- 206010073071 hepatocellular carcinoma Diseases 0.000 description 1
- 239000000833 heterodimer Substances 0.000 description 1
- 238000004128 high performance liquid chromatography Methods 0.000 description 1
- 239000004030 hiv protease inhibitor Substances 0.000 description 1
- 229940088597 hormone Drugs 0.000 description 1
- 239000005556 hormone Substances 0.000 description 1
- 102000046768 human CCL2 Human genes 0.000 description 1
- 210000005260 human cell Anatomy 0.000 description 1
- 244000052637 human pathogen Species 0.000 description 1
- 210000004408 hybridoma Anatomy 0.000 description 1
- 229910052739 hydrogen Inorganic materials 0.000 description 1
- 239000001257 hydrogen Substances 0.000 description 1
- 125000004435 hydrogen atom Chemical group [H]* 0.000 description 1
- 238000004191 hydrophobic interaction chromatography Methods 0.000 description 1
- GEZBIZMDLMTFDB-KJKHOEIASA-N hygrolidin Chemical compound CO[C@H]1\C=C\C=C(C)\C[C@H](C)[C@H](O)[C@H](C)\C=C(/C)\C=C(C)\C(=O)O[C@@H]1[C@@H](C)[C@@H](O)[C@H](C)[C@]1(O)O[C@H](C)[C@H](C)[C@H](OC(=O)\C=C\C(O)=O)C1 GEZBIZMDLMTFDB-KJKHOEIASA-N 0.000 description 1
- 206010020718 hyperplasia Diseases 0.000 description 1
- SSNQAUBBJYCSMY-KNTMUCJRSA-N hypothemycin Chemical compound O([C@@H](C)C\C=C/C(=O)[C@@H](O)[C@@H](O)C[C@H]1O[C@@H]11)C(=O)C=2C1=CC(OC)=CC=2O SSNQAUBBJYCSMY-KNTMUCJRSA-N 0.000 description 1
- 238000003384 imaging method Methods 0.000 description 1
- 230000002519 immonomodulatory effect Effects 0.000 description 1
- 210000000987 immune system Anatomy 0.000 description 1
- 238000003119 immunoblot Methods 0.000 description 1
- 230000036046 immunoreaction Effects 0.000 description 1
- 230000003308 immunostimulating effect Effects 0.000 description 1
- 230000001506 immunosuppresive effect Effects 0.000 description 1
- 229940124589 immunosuppressive drug Drugs 0.000 description 1
- 238000002650 immunosuppressive therapy Methods 0.000 description 1
- 230000002779 inactivation Effects 0.000 description 1
- 230000006698 induction Effects 0.000 description 1
- 206010022000 influenza Diseases 0.000 description 1
- 230000000977 initiatory effect Effects 0.000 description 1
- 150000002484 inorganic compounds Chemical class 0.000 description 1
- 229910010272 inorganic material Inorganic materials 0.000 description 1
- 238000003780 insertion Methods 0.000 description 1
- 230000037431 insertion Effects 0.000 description 1
- 230000010354 integration Effects 0.000 description 1
- 208000036971 interstitial lung disease 2 Diseases 0.000 description 1
- XMBWDFGMSWQBCA-YPZZEJLDSA-N iodane Chemical compound [125IH] XMBWDFGMSWQBCA-YPZZEJLDSA-N 0.000 description 1
- 229940044173 iodine-125 Drugs 0.000 description 1
- 238000006317 isomerization reaction Methods 0.000 description 1
- FABUFPQFXZVHFB-CFWQTKTJSA-N ixabepilone Chemical compound C/C([C@@H]1C[C@@H]2O[C@]2(C)CCC[C@@H]([C@@H]([C@H](C)C(=O)C(C)(C)[C@H](O)CC(=O)N1)O)C)=C\C1=CSC(C)=N1 FABUFPQFXZVHFB-CFWQTKTJSA-N 0.000 description 1
- 229960002014 ixabepilone Drugs 0.000 description 1
- 229960000318 kanamycin Drugs 0.000 description 1
- 229930027917 kanamycin Natural products 0.000 description 1
- SBUJHOSQTJFQJX-NOAMYHISSA-N kanamycin Chemical compound O[C@@H]1[C@@H](O)[C@H](O)[C@@H](CN)O[C@@H]1O[C@H]1[C@H](O)[C@@H](O[C@@H]2[C@@H]([C@@H](N)[C@H](O)[C@@H](CO)O2)O)[C@H](N)C[C@@H]1N SBUJHOSQTJFQJX-NOAMYHISSA-N 0.000 description 1
- 229930182823 kanamycin A Natural products 0.000 description 1
- 208000017169 kidney disease Diseases 0.000 description 1
- DAQAKHDKYAWHCG-RWTHQLGUSA-N lactacystin Chemical compound CC(=O)N[C@H](C(O)=O)CSC(=O)[C@]1([C@@H](O)C(C)C)NC(=O)[C@H](C)[C@@H]1O DAQAKHDKYAWHCG-RWTHQLGUSA-N 0.000 description 1
- 230000021633 leukocyte mediated immunity Effects 0.000 description 1
- 208000019423 liver disease Diseases 0.000 description 1
- HWYHZTIRURJOHG-UHFFFAOYSA-N luminol Chemical group O=C1NNC(=O)C2=C1C(N)=CC=C2 HWYHZTIRURJOHG-UHFFFAOYSA-N 0.000 description 1
- 206010025135 lupus erythematosus Diseases 0.000 description 1
- 210000002751 lymph Anatomy 0.000 description 1
- 230000003692 lymphatic flow Effects 0.000 description 1
- 108700021021 mRNA Vaccine Proteins 0.000 description 1
- 229940126582 mRNA vaccine Drugs 0.000 description 1
- 239000011777 magnesium Substances 0.000 description 1
- 229910052749 magnesium Inorganic materials 0.000 description 1
- 230000008774 maternal effect Effects 0.000 description 1
- 230000013011 mating Effects 0.000 description 1
- 238000005259 measurement Methods 0.000 description 1
- 235000013372 meat Nutrition 0.000 description 1
- 210000004379 membrane Anatomy 0.000 description 1
- WWNXYRCJJRRWAQ-ALCXHWRFSA-N mensacarcin Chemical compound O=C([C@@]1(O)[C@H](O)[C@]23O[C@]3([C@@H]([C@@H]1C)O)C(=O)C1=CC=CC(OC)=C1[C@@H]2OC)[C@@H]1O[C@H]1C WWNXYRCJJRRWAQ-ALCXHWRFSA-N 0.000 description 1
- ANZJBCHSOXCCRQ-FKUXLPTCSA-N mertansine Chemical compound CO[C@@H]([C@@]1(O)C[C@H](OC(=O)N1)[C@@H](C)[C@@H]1O[C@@]1(C)[C@@H](OC(=O)[C@H](C)N(C)C(=O)CCS)CC(=O)N1C)\C=C\C=C(C)\CC2=CC(OC)=C(Cl)C1=C2 ANZJBCHSOXCCRQ-FKUXLPTCSA-N 0.000 description 1
- 239000002207 metabolite Substances 0.000 description 1
- HIMMZWQWBNOPJH-UHFFFAOYSA-N methyl 4-[4-(dimethylamino)-6-methyl-5-[6-methyl-5-[(6-methyl-5-oxo-2h-pyran-2-yl)oxy]oxan-2-yl]oxyoxan-2-yl]oxy-2-ethyl-2,5,7,10-tetrahydroxy-6,11-dioxo-3,4-dihydro-1h-tetracene-1-carboxylate Chemical compound C12=C(O)C=3C(=O)C4=C(O)C=CC(O)=C4C(=O)C=3C=C2C(C(=O)OC)C(CC)(O)CC1OC(OC1C)CC(N(C)C)C1OC(OC1C)CCC1OC1OC(C)C(=O)C=C1 HIMMZWQWBNOPJH-UHFFFAOYSA-N 0.000 description 1
- 108091070501 miRNA Proteins 0.000 description 1
- HPNSFSBZBAHARI-UHFFFAOYSA-N micophenolic acid Natural products OC1=C(CC=C(C)CCC(O)=O)C(OC)=C(C)C2=C1C(=O)OC2 HPNSFSBZBAHARI-UHFFFAOYSA-N 0.000 description 1
- 238000001471 micro-filtration Methods 0.000 description 1
- 239000002679 microRNA Substances 0.000 description 1
- 108010032420 microcolin B Proteins 0.000 description 1
- 108010067094 microcystin Proteins 0.000 description 1
- 238000000386 microscopy Methods 0.000 description 1
- 230000003278 mimic effect Effects 0.000 description 1
- 229960004857 mitomycin Drugs 0.000 description 1
- KKZJGLLVHKMTCM-UHFFFAOYSA-N mitoxantrone Chemical compound O=C1C2=C(O)C=CC(O)=C2C(=O)C2=C1C(NCCNCCO)=CC=C2NCCNCCO KKZJGLLVHKMTCM-UHFFFAOYSA-N 0.000 description 1
- 229960001156 mitoxantrone Drugs 0.000 description 1
- 238000002156 mixing Methods 0.000 description 1
- 238000010369 molecular cloning Methods 0.000 description 1
- 239000000178 monomer Substances 0.000 description 1
- 210000003097 mucus Anatomy 0.000 description 1
- 230000036457 multidrug resistance Effects 0.000 description 1
- 201000006417 multiple sclerosis Diseases 0.000 description 1
- MINDHVHHQZYEEK-HBBNESRFSA-N mupirocin Chemical compound C[C@H](O)[C@H](C)[C@@H]1O[C@H]1C[C@@H]1[C@@H](O)[C@@H](O)[C@H](C\C(C)=C\C(=O)OCCCCCCCCC(O)=O)OC1 MINDHVHHQZYEEK-HBBNESRFSA-N 0.000 description 1
- 108010074166 muscotoxin A Proteins 0.000 description 1
- 230000000869 mutational effect Effects 0.000 description 1
- 229960000951 mycophenolic acid Drugs 0.000 description 1
- HPNSFSBZBAHARI-RUDMXATFSA-N mycophenolic acid Chemical compound OC1=C(C\C=C(/C)CCC(O)=O)C(OC)=C(C)C2=C1C(=O)OC2 HPNSFSBZBAHARI-RUDMXATFSA-N 0.000 description 1
- LXIQXFWXXPJPEE-NEYDLDDESA-N mytoxin b Chemical compound C([C@@]12[C@@]3(C)[C@@]45CCC(C)=C[C@H]4O[C@@H]1C[C@H]3OC(=O)/C=C/CC[C@@]1(OCC\C(=C/C(=O)OC5)[C@H]1O)C(=O)C)O2 LXIQXFWXXPJPEE-NEYDLDDESA-N 0.000 description 1
- ZTLGJPIZUOVDMT-UHFFFAOYSA-N n,n-dichlorotriazin-4-amine Chemical compound ClN(Cl)C1=CC=NN=N1 ZTLGJPIZUOVDMT-UHFFFAOYSA-N 0.000 description 1
- CMEGANPVAXDBPL-INIZCTEOSA-N n-[(7s)-1,2,3-trimethoxy-10-methylsulfanyl-9-oxo-6,7-dihydro-5h-benzo[a]heptalen-7-yl]acetamide Chemical compound C1([C@@H](NC(C)=O)CC2)=CC(=O)C(SC)=CC=C1C1=C2C=C(OC)C(OC)=C1OC CMEGANPVAXDBPL-INIZCTEOSA-N 0.000 description 1
- 239000002105 nanoparticle Substances 0.000 description 1
- 210000000822 natural killer cell Anatomy 0.000 description 1
- 230000017074 necrotic cell death Effects 0.000 description 1
- CTMCWCONSULRHO-UHQPFXKFSA-N nemorubicin Chemical compound C1CO[C@H](OC)CN1[C@@H]1[C@H](O)[C@H](C)O[C@@H](O[C@@H]2C3=C(O)C=4C(=O)C5=C(OC)C=CC=C5C(=O)C=4C(O)=C3C[C@](O)(C2)C(=O)CO)C1 CTMCWCONSULRHO-UHQPFXKFSA-N 0.000 description 1
- 229950010159 nemorubicin Drugs 0.000 description 1
- 108010068617 neonatal Fc receptor Proteins 0.000 description 1
- 230000003448 neutrophilic effect Effects 0.000 description 1
- RJMUSRYZPJIFPJ-UHFFFAOYSA-N niclosamide Chemical compound OC1=CC=C(Cl)C=C1C(=O)NC1=CC=C([N+]([O-])=O)C=C1Cl RJMUSRYZPJIFPJ-UHFFFAOYSA-N 0.000 description 1
- 229960001920 niclosamide Drugs 0.000 description 1
- 229920001220 nitrocellulos Polymers 0.000 description 1
- 239000002777 nucleoside Substances 0.000 description 1
- 125000003835 nucleoside group Chemical group 0.000 description 1
- KQZIMLFKBWIOJJ-CHNXQSITSA-N okilactomycin Chemical compound C[C@H]1C[C@@H](C)CCC[C@@H]2C=C(C(O)=O)[C@H](C)C[C@]32OC(=O)[C@]2(C)C(=O)C(=C)[C@H]1O[C@H]32 KQZIMLFKBWIOJJ-CHNXQSITSA-N 0.000 description 1
- 229930191479 oligomycin Natural products 0.000 description 1
- MNULEGDCPYONBU-AWJDAWNUSA-N oligomycin A Polymers O([C@H]1CC[C@H](/C=C/C=C/C[C@@H](C)[C@H](O)[C@@](C)(O)C(=O)[C@@H](C)[C@H](O)[C@@H](C)C(=O)[C@@H](C)[C@H](O)[C@@H](C)/C=C/C(=O)O[C@@H]([C@@H]2C)[C@@H]1C)CC)[C@@]12CC[C@H](C)[C@H](C[C@@H](C)O)O1 MNULEGDCPYONBU-AWJDAWNUSA-N 0.000 description 1
- 239000002751 oligonucleotide probe Substances 0.000 description 1
- 230000036407 pain Effects 0.000 description 1
- 239000002245 particle Substances 0.000 description 1
- 230000037361 pathway Effects 0.000 description 1
- 230000006320 pegylation Effects 0.000 description 1
- 229940111202 pepsin Drugs 0.000 description 1
- 230000001175 peptic effect Effects 0.000 description 1
- 238000010647 peptide synthesis reaction Methods 0.000 description 1
- 230000010412 perfusion Effects 0.000 description 1
- 238000002823 phage display Methods 0.000 description 1
- 210000001539 phagocyte Anatomy 0.000 description 1
- KUBDTFZQCYLLGC-VZORSVKHSA-N phallacidin Chemical compound N1C(=O)[C@@H]([C@@H](O)C(O)=O)NC(=O)[C@H](C(C)C)NC(=O)[C@H](C[C@@](C)(O)CO)NC(=O)[C@H](C2)NC(=O)[C@H](C)NC(=O)[C@@H]3C[C@H](O)CN3C(=O)[C@@H]1CSC1=C2C2=CC=CC=C2N1 KUBDTFZQCYLLGC-VZORSVKHSA-N 0.000 description 1
- CWCMIVBLVUHDHK-ZSNHEYEWSA-N phleomycin D1 Chemical compound N([C@H](C(=O)N[C@H](C)[C@@H](O)[C@H](C)C(=O)N[C@@H]([C@H](O)C)C(=O)NCCC=1SC[C@@H](N=1)C=1SC=C(N=1)C(=O)NCCCCNC(N)=N)[C@@H](O[C@H]1[C@H]([C@@H](O)[C@H](O)[C@H](CO)O1)O[C@@H]1[C@H]([C@@H](OC(N)=O)[C@H](O)[C@@H](CO)O1)O)C=1N=CNC=1)C(=O)C1=NC([C@H](CC(N)=O)NC[C@H](N)C(N)=O)=NC(N)=C1C CWCMIVBLVUHDHK-ZSNHEYEWSA-N 0.000 description 1
- NBIIXXVUZAFLBC-UHFFFAOYSA-K phosphate Chemical compound [O-]P([O-])([O-])=O NBIIXXVUZAFLBC-UHFFFAOYSA-K 0.000 description 1
- 239000010452 phosphate Substances 0.000 description 1
- 229940097886 phosphorus 32 Drugs 0.000 description 1
- 230000000704 physical effect Effects 0.000 description 1
- 230000036417 physical growth Effects 0.000 description 1
- BBLGCDSLCDDALX-LKGBESRRSA-N piericidin A Chemical compound COC=1NC(C\C=C(/C)C\C=C\C(\C)=C\[C@@H](C)[C@@H](O)C(\C)=C\C)=C(C)C(=O)C=1OC BBLGCDSLCDDALX-LKGBESRRSA-N 0.000 description 1
- 229920000867 polyelectrolyte Polymers 0.000 description 1
- OWRCRVABZYJUES-FLJLGRLJSA-N polyketomycin Polymers O([C@H]1[C@](C)(O)C[C@@H](O[C@H]1C)O[C@H]1CC[C@@H](O[C@@H]1C)O[C@@H]1C(=O)C(=C(/C)O)/C(=O)[C@@]2(O)C(=O)C=3C(O)=C4C(=O)C=C(C(C4=C(C)C=3C[C@]21O)=O)OC)C(=O)C1=C(C)C=CC(C)=C1O OWRCRVABZYJUES-FLJLGRLJSA-N 0.000 description 1
- 229920000642 polymer Polymers 0.000 description 1
- 229920002451 polyvinyl alcohol Polymers 0.000 description 1
- 239000013641 positive control Substances 0.000 description 1
- 230000001323 posttranslational effect Effects 0.000 description 1
- 238000001556 precipitation Methods 0.000 description 1
- 229960004618 prednisone Drugs 0.000 description 1
- XOFYZVNMUHMLCC-ZPOLXVRWSA-N prednisone Chemical compound O=C1C=C[C@]2(C)[C@H]3C(=O)C[C@](C)([C@@](CC4)(O)C(=O)CO)[C@@H]4[C@@H]3CCC2=C1 XOFYZVNMUHMLCC-ZPOLXVRWSA-N 0.000 description 1
- 238000002360 preparation method Methods 0.000 description 1
- 125000002924 primary amino group Chemical group [H]N([H])* 0.000 description 1
- 238000007639 printing Methods 0.000 description 1
- 210000001236 prokaryotic cell Anatomy 0.000 description 1
- 230000035755 proliferation Effects 0.000 description 1
- 238000011321 prophylaxis Methods 0.000 description 1
- 108020003519 protein disulfide isomerase Proteins 0.000 description 1
- 230000012846 protein folding Effects 0.000 description 1
- 238000001243 protein synthesis Methods 0.000 description 1
- SLYDIPAXCVVRNY-UOWMTANKSA-N pseurotin A Chemical compound O1C([C@@H](O)[C@@H](O)\C=C/CC)=C(C)C(=O)[C@]21C(=O)N[C@](C(=O)C=1C=CC=CC=1)(OC)[C@@H]2O SLYDIPAXCVVRNY-UOWMTANKSA-N 0.000 description 1
- BWXSGIGGLVLYDS-UHFFFAOYSA-N pseurotin A Natural products CCC=C/C(O)C(O)C1=CC(=O)C2(O1)C(O)C(NC2=O)(OC)C(=O)c3ccccc3 BWXSGIGGLVLYDS-UHFFFAOYSA-N 0.000 description 1
- 208000005069 pulmonary fibrosis Diseases 0.000 description 1
- 229950010131 puromycin Drugs 0.000 description 1
- YUOCYTRGANSSRY-UHFFFAOYSA-N pyrrolo[2,3-i][1,2]benzodiazepine Chemical compound C1=CN=NC2=C3C=CN=C3C=CC2=C1 YUOCYTRGANSSRY-UHFFFAOYSA-N 0.000 description 1
- 108700023331 quinaldopeptin Proteins 0.000 description 1
- 229960005567 rebeccamycin Drugs 0.000 description 1
- INSACQSBHKIWNS-QZQSLCQPSA-N rebeccamycin Chemical compound O[C@@H]1[C@@H](O)[C@H](OC)[C@@H](CO)O[C@H]1N1C2=C3N=C4[C](Cl)C=CC=C4C3=C3C(=O)NC(=O)C3=C2C2=CC=CC(Cl)=C21 INSACQSBHKIWNS-QZQSLCQPSA-N 0.000 description 1
- 239000002464 receptor antagonist Substances 0.000 description 1
- 229940044551 receptor antagonist Drugs 0.000 description 1
- 108091006082 receptor inhibitors Proteins 0.000 description 1
- 238000011084 recovery Methods 0.000 description 1
- 230000003362 replicative effect Effects 0.000 description 1
- 238000011160 research Methods 0.000 description 1
- 108091008146 restriction endonucleases Proteins 0.000 description 1
- PYWVYCXTNDRMGF-UHFFFAOYSA-N rhodamine B Chemical compound [Cl-].C=12C=CC(=[N+](CC)CC)C=C2OC2=CC(N(CC)CC)=CC=C2C=1C1=CC=CC=C1C(O)=O PYWVYCXTNDRMGF-UHFFFAOYSA-N 0.000 description 1
- 239000003730 rna directed rna polymerase inhibitor Substances 0.000 description 1
- 150000003839 salts Chemical class 0.000 description 1
- 238000005070 sampling Methods 0.000 description 1
- 108700026174 sandramycin Proteins 0.000 description 1
- WXIVYIYCEBUEHL-RTQGILJWSA-N sandramycin Chemical compound C([C@H](C(=O)N1CCCC[C@H]1C(=O)NCC(=O)N(C)CC(=O)N(C)[C@H](C(OC1)=O)C(C)C)NC(=O)C=2C(=CC3=CC=CC=C3N=2)O)OC(=O)[C@H](C(C)C)N(C)C(=O)CN(C)C(=O)CNC(=O)[C@@H]2CCCCN2C(=O)[C@@H]1NC(=O)C1=NC2=CC=CC=C2C=C1O WXIVYIYCEBUEHL-RTQGILJWSA-N 0.000 description 1
- 229940084560 sanguinarine Drugs 0.000 description 1
- YZRQUTZNTDAYPJ-UHFFFAOYSA-N sanguinarine pseudobase Natural products C1=C2OCOC2=CC2=C3N(C)C(O)C4=C(OCO5)C5=CC=C4C3=CC=C21 YZRQUTZNTDAYPJ-UHFFFAOYSA-N 0.000 description 1
- 210000000717 sertoli cell Anatomy 0.000 description 1
- 238000004904 shortening Methods 0.000 description 1
- 229950008974 sinefungin Drugs 0.000 description 1
- 238000009097 single-agent therapy Methods 0.000 description 1
- 239000004055 small Interfering RNA Substances 0.000 description 1
- 230000000391 smoking effect Effects 0.000 description 1
- 239000011780 sodium chloride Substances 0.000 description 1
- 239000000243 solution Substances 0.000 description 1
- 238000004611 spectroscopical analysis Methods 0.000 description 1
- 238000007619 statistical method Methods 0.000 description 1
- 229910052717 sulfur Inorganic materials 0.000 description 1
- 230000000153 supplemental effect Effects 0.000 description 1
- 238000002198 surface plasmon resonance spectroscopy Methods 0.000 description 1
- 238000013268 sustained release Methods 0.000 description 1
- 239000012730 sustained-release form Substances 0.000 description 1
- 238000003786 synthesis reaction Methods 0.000 description 1
- 229950009150 taltobulin Drugs 0.000 description 1
- YVSQVYZBDXIXCC-INIZCTEOSA-N telomestatin Chemical compound N=1C2=COC=1C(N=1)=COC=1C(N=1)=COC=1C(N=1)=COC=1C(N=1)=COC=1C(=C(O1)C)N=C1C(=C(O1)C)N=C1[C@@]1([H])N=C2SC1 YVSQVYZBDXIXCC-INIZCTEOSA-N 0.000 description 1
- 229960002180 tetracycline Drugs 0.000 description 1
- 229930101283 tetracycline Natural products 0.000 description 1
- 235000019364 tetracycline Nutrition 0.000 description 1
- 150000003522 tetracyclines Chemical class 0.000 description 1
- 125000003396 thiol group Chemical group [H]S* 0.000 description 1
- 210000000115 thoracic cavity Anatomy 0.000 description 1
- 229940104230 thymidine Drugs 0.000 description 1
- 239000003104 tissue culture media Substances 0.000 description 1
- 229960003989 tocilizumab Drugs 0.000 description 1
- 239000011573 trace mineral Substances 0.000 description 1
- 235000013619 trace mineral Nutrition 0.000 description 1
- 108091006106 transcriptional activators Proteins 0.000 description 1
- 238000012546 transfer Methods 0.000 description 1
- 238000002054 transplantation Methods 0.000 description 1
- YKUJZZHGTWVWHA-UHFFFAOYSA-N triptolide Natural products COC12CC3OC3(C(C)C)C(O)C14OC4CC5C6=C(CCC25C)C(=O)OC6 YKUJZZHGTWVWHA-UHFFFAOYSA-N 0.000 description 1
- 229910052722 tritium Inorganic materials 0.000 description 1
- GOVYBPLHWIEHEJ-UHFFFAOYSA-N tubastatin A Chemical compound C1N(C)CCC2=C1C1=CC=CC=C1N2CC1=CC=C(C(=O)NO)C=C1 GOVYBPLHWIEHEJ-UHFFFAOYSA-N 0.000 description 1
- 201000008827 tuberculosis Diseases 0.000 description 1
- 229930184737 tubulysin Natural products 0.000 description 1
- 210000004881 tumor cell Anatomy 0.000 description 1
- 210000001944 turbinate Anatomy 0.000 description 1
- 108010087967 type I signal peptidase Proteins 0.000 description 1
- 230000036269 ulceration Effects 0.000 description 1
- 238000005199 ultracentrifugation Methods 0.000 description 1
- ORHBXUUXSCNDEV-UHFFFAOYSA-N umbelliferone Chemical compound C1=CC(=O)OC2=CC(O)=CC=C21 ORHBXUUXSCNDEV-UHFFFAOYSA-N 0.000 description 1
- HFTAFOQKODTIJY-UHFFFAOYSA-N umbelliferone Natural products Cc1cc2C=CC(=O)Oc2cc1OCC=CC(C)(C)O HFTAFOQKODTIJY-UHFFFAOYSA-N 0.000 description 1
- 238000009424 underpinning Methods 0.000 description 1
- 108020005087 unfolded proteins Proteins 0.000 description 1
- 241000712461 unidentified influenza virus Species 0.000 description 1
- 241001430294 unidentified retrovirus Species 0.000 description 1
- 210000002700 urine Anatomy 0.000 description 1
- VBEQCZHXXJYVRD-GACYYNSASA-N uroanthelone Chemical compound C([C@@H](C(=O)N[C@H](C(=O)N[C@@H](CS)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CS)C(=O)N[C@H](C(=O)N[C@@H]([C@@H](C)CC)C(=O)NCC(=O)N[C@@H](CC=1C=CC(O)=CC=1)C(=O)N[C@@H](CO)C(=O)NCC(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CS)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCCNC(N)=N)C(O)=O)C(C)C)[C@@H](C)O)NC(=O)[C@H](CO)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CO)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@@H](NC(=O)[C@H](CC=1NC=NC=1)NC(=O)[C@H](CCSC)NC(=O)[C@H](CS)NC(=O)[C@@H](NC(=O)CNC(=O)CNC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CS)NC(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)CNC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)[C@H](CO)NC(=O)[C@H](CO)NC(=O)[C@H]1N(CCC1)C(=O)[C@H](CS)NC(=O)CNC(=O)[C@H]1N(CCC1)C(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)[C@H](CO)NC(=O)[C@@H](N)CC(N)=O)C(C)C)[C@@H](C)CC)C1=CC=C(O)C=C1 VBEQCZHXXJYVRD-GACYYNSASA-N 0.000 description 1
- 239000004474 valine Substances 0.000 description 1
- ZWCXYZRRTRDGQE-LUPIJMBPSA-N valyl gramicidin a Chemical compound C1=CC=C2C(C[C@H](NC(=O)[C@@H](CC(C)C)NC(=O)[C@H](CC=3C4=CC=CC=C4NC=3)NC(=O)[C@@H](CC(C)C)NC(=O)[C@H](CC=3C4=CC=CC=C4NC=3)NC(=O)[C@@H](CC(C)C)NC(=O)[C@H](CC=3C4=CC=CC=C4NC=3)NC(=O)[C@@H](C(C)C)NC(=O)[C@H](C(C)C)NC(=O)[C@@H](C(C)C)NC(=O)[C@H](C)NC(=O)[C@H](NC(=O)[C@H](C)NC(=O)CNC(=O)[C@@H](NC=O)C(C)C)CC(C)C)C(=O)NCCO)=CNC2=C1 ZWCXYZRRTRDGQE-LUPIJMBPSA-N 0.000 description 1
- 230000002792 vascular Effects 0.000 description 1
- 238000009423 ventilation Methods 0.000 description 1
- 230000003519 ventilatory effect Effects 0.000 description 1
- 230000035899 viability Effects 0.000 description 1
- 230000009385 viral infection Effects 0.000 description 1
- 230000000007 visual effect Effects 0.000 description 1
- 229920003169 water-soluble polymer Polymers 0.000 description 1
- 230000004584 weight gain Effects 0.000 description 1
- 235000019786 weight gain Nutrition 0.000 description 1
- JFCFGYGEYRIEBE-YVLHJLIDSA-N wob38vs2ni Chemical compound CO[C@@H]([C@@]1(O)C[C@H](OC(=O)N1)[C@@H](C)[C@@H]1O[C@@]1(C)[C@@H](OC(=O)[C@H](C)N(C)C(=O)CCC(C)(C)S)CC(=O)N1C)\C=C\C=C(C)\CC2=CC(OC)=C(Cl)C1=C2 JFCFGYGEYRIEBE-YVLHJLIDSA-N 0.000 description 1
- 238000002424 x-ray crystallography Methods 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P11/00—Drugs for disorders of the respiratory system
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/395—Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
-
- 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
-
- 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]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/505—Medicinal preparations containing antigens or antibodies comprising antibodies
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/30—Immunoglobulins specific features characterized by aspects of specificity or valency
- C07K2317/33—Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/70—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 disclosure generally pertains to antibodies and antigen-binding fragments thereof, preferably human antibodies and antigen-binding fragments and/or affinity-matured variants thereof, recombinant cells engineered to express such antibodies, and compositions containing such antibodies and antigen-binding fragments thereof, wherein such antibodies and antigen-binding fragments thereof specifically bind to the S protein of coronaviruses (“CoV-S”) and therapeutic and diagnostic uses for the antibodies, antigen-binding fragments, and compositions thereof.
- CoV-S coronaviruses
- Coronaviruses (“CoV”) are genetically classified into four major genera: the Alphacoronavirus genus (ACoV genus); the Betacoronavirus genus (BCoV genus); the Gammacoronavirus genus (CCoV genus); and Deltacoronavirus genus (DCoV genus), and while ACoV and BCoV primarily infect a als CCoV and DCoV predominantly infect birds (Wu A. et al, Cell Host Microbe. 2020 Mar ll;27(3):325-328). Coronaviruses that infect humans were first identified in the mid-1960s, and currently, seven confirmed CoV species are known as human pathogens.
- SARS Severe Acute Respiratory Syndrome
- MERS-CoV Middle East Respiratory Syndrome
- 2019-nCoV-2 also known as “2019-nCoV”
- the genome of coronaviruses includes a variable number (from 6 to 11) of open reading frames (“ORFs”) (Wu A. et al. , Cell Host Microbe. 2020 Mar ll;27(3):325-328).
- ORFs open reading frames
- the first ORF encodes 16 non-structural proteins (“nsps”), and the remaining ORFs encode accessory proteins and structural proteins.
- the four major structural proteins are the spike surface glycoprotein (“S protein” or “S” or “spike protein”), small envelope protein (“E protein” or “E”), matrix protein (“M protein” or “M”), and nucleocapsid protein (“N protein”, or “N”).
- the S protein which plays an essential role in binding to receptors on the host cell and determines host tropism (Zhu Z. et al, Infect Genet Evol. 2018 Jul;61 : 183-184), forms homotrimers protruding from the viral surface (Li F. Annu Rev Virol. 2016 Sep 29;3(1):237-261).
- the S protein is processed into two non-covalently associated subunits, SI and S2, and each monomer in the trimeric S assembly is a heterodimer of SI and S2 subunits.
- NTD N-terminal domain
- CCD C-terminal domain
- the CTD functions as the receptor-binding domain (RBD) for both SARS-CoV and SARS-CoV-2 (Li F. J Virol. 2015 Feb;89(4): 1954-64).
- the S2 subunit contains the fusion peptide, heptad repeat 1 and 2, and a transmembrane domain, all of which are required to mediate fusion of the viral and host cell membranes.
- SARS-CoV and SARS-CoV-2 bind to and use angiotensin-converting enzyme 2 (ACE2) of a host cell as a receptor to enter the host cells (Ge X.Y. et al, Nature. 2013 Nov 28;503(7477):535-8; Hoffmann M. et al, Cell. 2020 Mar 4. pii: S0092-8674(20)30229-4).
- ACE2 angiotensin-converting enzyme 2
- the motif within the RBD that particularly binds to RCE2 is often referred to as the “ACE2-binding motif’.
- SARS-CoV can also use CD209L (also known as L-SIGN) as an alternative receptor (Jeffers S. A. et al, Proc Natl Acad Sci U S A. 2004 Nov 2;101(44):15748-53).
- MERS-CoV binds dipeptidyl peptidase 4 (“DPP4”, also known as CD26) of the host cell via a
- SARS-CoV-2 The genome of SARS-CoV-2 is about 29.8 kb nucleotides and encodes 15 nsps, four structural proteins (S, E, M, and N) and eight accessory proteins (3a, 3b, p6, 7a, 7b, 8b, 9b, and orfl4) (Wu A. et al, Cell Host Microbe. 2020 Mar ll;27(3):325-328). While SARS-CoV-2 is genetically close to a SARS-like bat CoV and also to SARS-CoV, a number of sequence differences have been identified.
- SARS-CoV-2 When SARS-CoV-2 is compared to SARS-CoV or SARS-like bat CoV, 380 amino acid differences or substitutions were found, 27 of which are in the S protein, including 6 substitutions in the RBD at amino acid region 357-528 (but not in the receptor-binding motifs that directly interact with ACE2) and 6 substitutions in the underpinning subdomain (SD) at amino acid region 569-655.
- FDA U.S. Food and Drug Administration
- remdesivir One of the few drugs approved by the U.S. Food and Drug Administration (“FDA”) for use in treating COVID-19 is the viral replication inhibitor remdesivir. Clinical trials demonstrated that remdesivir shortens the time to recovery in hospitalized patients, but more effective therapy is in great need. Convalescent plasma received the emergency use authorization status by the FDA.
- lopinavir-ritonavir HIV protease inhibitor
- ABX464 viral RNA splicer
- favilavir RNA-dependent RNA polymerase inhibitor used for influenza virus infection
- niclosamide and ivermectin antihelmintic
- BCG vaccine vaccine for tuberculosis
- IL-6 receptor antagonist antibodies an anti-GM-CSF or anti-GM-CSF receptor antibody, an anti-TNF antibody, an anti-IL- lbeta antibody, or an anti-complement component 5 antibody, in an effort to inhibit inflammation and thereby potentially inhibit cytokine storm and sepsis which can manifest in some SARS-CoV-2- infected patients and may cause death.
- the present disclosure relates to a compound which binds to coronavirus (CoV) or the spike protein (S protein) of a CoV (“CoV-S”).
- the compound may be an isolated antibody or antigen-binding antibody fragment which binds to a CoV-S.
- the antibody or antigen-binding antibody fragment may comprise a heavy chain variable region (VH), or fragments thereof, and/or a light chain variable region (VL), or fragments thereof.
- the VH or fragment thereof may comprise a complementarity determining region 1 (CDR1), a complementarity-determining region 2 (CDR2), and a complementarity-determining region 3 (CDR3), which may also be referred to as VH CDR1, VH CDR2, and VH CDR3, respectively.
- the VL or fragment thereof may comprise a CDR1, a CDR2, and a CDR3, which may also be referred to as VL CDR1, VL CDR2, and VL CDR3, respectively.
- the antibody, or antigen-binding antibody fragment thereof may comprise a heavy chain CDR1, a heavy chain CDR2, a heavy chain CDR3, a light chain CDR1, a light chain CDR2, and a light chain CDR3.
- the antibody or antigen-binding antibody fragment may comprise an antibody or antigen-binding antibody fragment thereof, or an affinity-matured variant of an anti-Co V- S antibody or antigen-binding antibody fragment thereof; selected from the group consisting of ADI- 58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI- 58128, ADI-58129, ADI-58130, ADI-58131, optionally wherein the CoV-S is SARS-CoV-S or SARS-CoV-2-S.
- the antibody, or antigen-binding antibody fragment thereof may comprise a VH and/or VL.
- the VH may comprise a CDR3 having an amino acid sequence identical to the VH CDR3 of any one of anti-CoV-S antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, ADI-58131
- the VL CDR3 may comprise a CDR3 having an amino acid sequence identical to the VL CDR3 of the same anti-CoV-S antibody that the VH CDR3 is derived from, and the anti-CoV-S antibody may be selected from any one of anti-CoV-S antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI- 58122, ADI
- the CoV-S may be the spike protein (“S protein”) of Severe Acute Respiratory Syndrome (SARS) coronavirus (“SARS-CoV”), which may be referred to as “SARS- CoV-S”, or the S protein of SARS-CoV -2 (also known as “n2019-nCoV”), which may be referred to as “SARS-CoV -2-S”.
- S protein spike protein
- SARS-CoV-S Severe Acute Respiratory Syndrome coronavirus
- n2019-nCoV also known as “n2019-nCoV”
- the CoV-S may comprise a sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% identity to, comprising, or consisting of the amino acid sequence of SEQ ID NO: 401 (SARS-CoV-S, 1288 amino acids, Accession# PDB: 6VSB_B) or having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% identity to, comprising, or consisting of SEQ ID NO: 403 (SARS-CoV -2-S, 1273 amino acids, GenBank: QHD43416.1).
- the SARS-CoV -2-S is a B.1.1.7 variant, a B. 1.351 variant, a B.1.1.28 variant, a B. 1.429 variant, a P.l varaint, a B.1.617 variant (e.g ., B.1.617.1 and B.1.617.2), a C.37 variant, a 1.621 variant, a AY.l variant, a 1.623 variant, a C.36 variant, a A.27 variant, a AV.l variant, a B.1.1.482 variant, a B.1.1.523 variant, a B.1.427 variant, a AY.4 variant, a AY.ll variant, variant, a D614G variant of SEQ ID NO: 403, a B.1.1.529/BA.1 variant (also known as the Omicron variant), a BA.1.1 variant, or a BA.2 variant.
- a B.1.1.529/BA.1 variant also known as the Omicron variant
- the antibody or antigen-binding antibody fragment may comprise at least 1, 2, 3, 4, 5 or all 6 complementarity-determining regions (CDRs) of any one of anti-CoV-S antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, ADI-58131, optionally wherein the CoV-S is SARS-CoV-S or SARS-CoV-2-S.
- CDRs complementarity-determining regions
- the CoV-S may comprise the amino acid sequence of SEQ ID NO: 401 (SARS-CoV-S, 1288 amino acids, Accession# PDB: 6VSB_B) or SEQ ID NO: 403 (SARS-CoV-2-S, 1273 amino acids, GenBank: QHD43416.1).
- the isolated antibody or antigen-binding antibody fragment may comprise: (a) a VH CDR1 polypeptide; (b) a VH CDR2 polypeptide; (c) a VH CDR3 polypeptide; (d) a VL CDR1 polypeptide; (e) a VL CDR2 polypeptide; and (f) a VL CDR3 polypeptide.
- amino acid sequences of the VH CDR1, the VH CDR2, the VH CDR3, the VL CDR1, the VL CDR2, and the VL CDR3 may be identical to the amino acid sequences of the VH CDR1, VH CDR2, VH CDR3, VL CDR1,
- the CoV-S may be SARS- CoV-S or of “SARS-CoV-2-S”.
- the CoV-S may comprise the amino acid sequence of SEQ ID NO: 401 (SARS-CoV-S, 1288 amino acids, Accession# PDB: 6VSB_B) or SEQ ID NO: 403 (SARS-CoV-2-S, 1273 amino acids, GenBank: QHD43416.1).
- the isolated antibody or antigen-binding antibody fragment may comprise: (a) a VH comprising a VH CDR1, VH CDR2, and VH CDR3; and (b) a VL comprising a VL CDR1, VL CDR2, and VL CDR3.
- the amino acid sequences of the VH CDR1, the VH CDR2, the VH CDR3, the VL CDR1, the VL CDR2, and the VL CDR3 may be identical to the amino acid sequences of: (1) SEQ ID NOS: 2, 4, 6, 202, 204, and 206, respectively; (2) SEQ ID NOS: 12, 14, 16, 212, 214, and 216, respectively; (3) SEQ ID NOS: 22, 24, 26, 222, 224, and 226, respectively; (4)
- the amino acid sequences of the VH CDR1, the VH CDR2, the VH CDR3, the VL CDR1, the VL CDR2, and the VL CDR3 may be identical to the VH CDR1, the VH CDR2, the VH CDR3, the VL CDR1, the VL CDR2, and the VL CDR3 and VL amino acid sequences of any one of anti-CoV-S antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, ADI-58131.
- the isolated antibody or antigen-binding antibody fragment may possess one of the following structural features:
- the VH may comprise an amino acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 8
- the VL may comprise an amino acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 208;
- the VH may comprise an amino acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 18, and (b) the VL may comprise an amino acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 218;
- the VH may comprise an amino acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 28, and (b) the VL may comprise an amino acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 228;
- the VH may comprise an amino acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 38
- the VL may comprise an amino acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 238;
- the VH may comprise an amino acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 48
- the VL may comprise an amino acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 248;
- the VH may comprise an amino acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 58
- the VL may comprise an amino acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 258;
- the VH may comprise an amino acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 68
- the VL may comprise an amino acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 268;
- the VH may comprise an amino acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 78
- the VL may comprise an amino acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 278;
- the VH may comprise an amino acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 88
- the VL may comprise an amino acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 288;
- the VH may comprise an amino acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 98
- the VL may comprise an amino acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 298;
- the VH may comprise an amino acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 108
- the VL may comprise an amino acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 308; or
- the VH may comprise an amino acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 118
- the VL may comprise an amino acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 318.
- the isolated antibody or antigen-binding antibody fragment may be human, humanized, primatized or chimeric.
- the isolated antibody or antigen-binding antibody fragment may be bispecific or multispecific.
- the isolated antibody or antigen-binding antibody fragment may comprise at least one first antigen-binding domain (“ABD”) and at least one second ABD.
- the first ABD may comprise the VH CDR1, the VH CDR2, the VH CDR3, the VL CDR1, the VL CDR2, and the VL CDR3 of a first anti-CoV-S antibody selected from any one of anti-CoV-S antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, ADI-58131; and/or
- the second ABD may comprise the VH CDR1, the VH CDR2, the VH CDR3, the VL CDR1, the VL CDR2, and the VL CDR3 of a second anti-CoV-S antibody selected from any one of anti-CoV-S antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, ADI-58131.
- the first anti-CoV-S antibody may be same as the second anti-CoV-S antibody or may be different from the second anti-CoV-S antibody.
- the first anti-CoV-S antibody and the second anti-CoV-S antibody may bind to the same or different coronavirus species.
- the first CoV-S and the second CoV-S may be (i) both of SARS-CoV or (ii) both of SARS-CoV-2.
- the first anti-CoV-S antibody may be same as the second anti-CoV-S antibody or may be different from the second anti-CoV-S antibody. Still further optionally, these antibodies may bind to the same or different epitopes on a CoV-S expressed by said SARS-CoV or SARS-CoV-2.
- the first anti-CoV-S antibody and the second anti-CoV-S antibody may bind to different coronaviruses, optionally wherein the first CoV-S and the second CoV-S are (i) SARS-CoV and of SARS-CoV-2 coronaviruses, respectively, or are (ii) SARS-CoV-2 and of SARS- CoV coronaviruses, respectively.
- the bispecific or multispecific isolated antibody or antigen-binding antibody fragment may comprise at least one first ABD and at least one second ABD.
- the first ABD may comprise the VH CDR1, the VH CDR2, the VH CDR3, the VL CDR1, the VL CDR2, and the VL CDR3 of a first anti-CoV-S antibody selected from any one of anti-CoV-S antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, ADI-58131, or an affinity-matured variant of any of the foregoing; and/or (b) the second ABD binds to an antigen which may not be a CoV-S, optionally wherein the antigen is a cytokine, a cytokine receptor, or an immunomodulatory polypeptide.
- the isolated antibody or antigen-binding antibody fragment may comprise a Fab, Fab’, F(ab’)2, scFv, sc(Fv)2, minibody, diabody, sdAb, BITE.
- the isolated antibody or antigen-binding antibody fragment may comprise a constant region or Fc region or at least one domain thereof.
- the constant region or Fc region may comprise a mutation which impairs or enhances at least one effector function, optionally FcR binding, FcRn binding, complement binding, glycosylation, complement-dependent cytotoxicity (“CDC”), or antibody-dependent cellular cytotoxicity (“ADCC”).
- effector function optionally FcR binding, FcRn binding, complement binding, glycosylation, complement-dependent cytotoxicity (“CDC”), or antibody-dependent cellular cytotoxicity (“ADCC”).
- the constant or Fc region is primate derived, preferably human.
- the human constant or Fc region optionally may be selected from a human IgGl, IgG2, IgG3 or IgG4 constant or Fc region which optionally may be modified, optionally such as by domain deletion or by introducing one or more mutations which impair or enhance at least one effector function.
- the present disclosure further relates to chimeric antigen receptors (“CARs”) comprising at least one antibody or antigen-binding antibody fragment described herein.
- ADCs antibody-drug conjugates
- ADCs comprising: (a) at least one antibody or antigen-binding antibody fragment described herein; and (b) a drug.
- the drug may be: (i) an antiviral drug, which is optionally, remdesivir, favipiravir, darunavir, nelfinavir, saquinavir, lopinavir or ritonavir; (ii) an antihelminth drug, which may be optionally ivermectin; (iii) an antiparasite drug, which may be optionally hydroxychloroquine, chloroquine, or atovaquone; (iv) antibacterial vaccine, which may be optionally the tuberculosis vaccine BCG; or (v) an anti-inflammatory drug, which may be optionally a steroid such as ciclesonide, a TNF inhibitor (e.g., adalimumab), a TNF receptor inhibitor (e.g., etanercept), an IL-6 inhibitor (e.g., clazakizumab), an IL-6 receptor inhibitor (e.g.,
- the present disclosure also relates to isolated nucleic acids encoding any of the antibodies or antigen-binding antibody fragments disclosed herein.
- the nucleic acid may comprise:
- nucleic acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the nucleic acid sequence of SEQ ID NO: 9, and/or (b) a nucleic acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the nucleic acid sequence of SEQ ID NO: 209; (2) (a) a nucleic acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the nucleic acid sequence of SEQ ID NO: 19, and/or (b) a nucleic acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the nucleic acid sequence of SEQ ID NO: 219;
- nucleic acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the nucleic acid sequence of SEQ ID NO: 29, and/or (b) a nucleic acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the nucleic acid sequence of SEQ ID NO: 229;
- nucleic acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the nucleic acid sequence of SEQ ID NO: 39, and/or (b) a nucleic acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the nucleic acid sequence of SEQ ID NO: 239;
- nucleic acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the nucleic acid sequence of SEQ ID NO: 49, and/or (b) a nucleic acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the nucleic acid sequence of SEQ ID NO: 249;
- nucleic acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the nucleic acid sequence of SEQ ID NO: 59, and/or (b) a nucleic acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the nucleic acid sequence of SEQ ID NO: 259;
- nucleic acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the nucleic acid sequence of SEQ ID NO: 69, and/or (b) a nucleic acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the nucleic acid sequence of SEQ ID NO: 269;
- nucleic acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the nucleic acid sequence of SEQ ID NO: 79, and/or (b) a nucleic acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the nucleic acid sequence of SEQ ID NO: 279;
- nucleic acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the nucleic acid sequence of SEQ ID NO: 89, and/or (b) a nucleic acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the nucleic acid sequence of SEQ ID NO: 289;
- nucleic acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the nucleic acid sequence of SEQ ID NO: 99, and/or (b) a nucleic acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the nucleic acid sequence of SEQ ID NO: 299; (11) (a) a nucleic acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the nucleic acid sequence of SEQ ID NO: 109, and/or (b) a nucleic acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the nucleic acid sequence of SEQ ID NO: 309; or
- the present disclosure also relates to isolated cells which may comprise any of the nucleic acids disclosed herein.
- the cell may be a bacterial, yeast, insect, fungal, or mammalian cell, optionally a human cell, further optionally a CHO or HEK cell.
- the cell may be a human immune cell, optionally a T, NK, B or dendritic cell.
- the present disclosure further relates to methods of expressing the antibody or antigen binding antibody fragment or the CAR disclosed herein.
- the method may comprise: (a) culturing the cell expressing an antibody or antigen-binding antibody fragment or CAR of the present disclosure under conditions that permit expression; and (b) optionally isolating the antibody or antigen-binding antibody fragment or the CAR from the cell or the culture medium containing the cell.
- the present disclosure further relates to methods of identifying an antibody or an antigen binding antibody fragment which specifically binds to CoV-S.
- the method may comprise: (a) obtaining antisera and/or B cells obtained from a patient infected with SARS-CoV or SARS-CoV-2, optionally wherein the patient recovered from SARS-CoV or SARS-CoV-2 infection or the patient is a convalescent patient infected with SARS-CoV or SARS-CoV-2; (b) contacting the antisera and/or B cells with the CoV-S; and (c) isolating an antibody or antigen-binding fragment thereof which specifically bind to the CoV-S.
- the CoV-S is the spike protein of SARS-CoV (“SARS-CoV-S”) or of SARS-CoV-2 (“SARS-CoV-2-S”).
- the CoV-S may comprise the amino acid sequence of SEQ ID NO: 401 (SARS-CoV-S, 1288 amino acids, Accession# PDB: 6VSB_B) or SEQ ID NO: 403 (SARS- CoV -2-S, 1273 amino acids, GenBank: QHD43416.1).
- the method may further detect that the antibody or antigen-binding fragment thereof which specifically binds to CoV-S neutralizes, blocks or inhibits coronavirus infectivity or coronavirus proliferation, optionally wherein the coronavirus is SARS-CoV or SARS- CoV-2.
- the method may further detect whether the antibody or antigen binding antibody fragment thereof which specifically binds to the CoV-S binds to other coronaviruses, optionally selected from the group consisting of MERS-CoV, HCoV-HKUl, HCoV- OC43, HCoV-229E, and HCoV-NL63.
- the method may further comprise determining the sequence of the antibody or antigen-binding antibody fragment thereof may be determined.
- these sequences may be affinity-matured or mutated to enhance binding affinity and/or potentially increase specificity to a particular CoV-S.
- compositions comprising: (a) at least one antibody or antigen-binding antibody fragment of the present disclosure; and (b) a pharmaceutically acceptable carrier or excipient.
- the present disclosure further provides methods of determining whether a subject has been infected with SARS-CoV or SARS-CoV-2 or another coronavirus by detecting whether a biological sample from the subject may comprise SARS-CoV-S protein or SARS-CoV-S-2 protein or another coronavirus S protein homologous thereto based on its immunoreaction with at least one antibody or antigen-binding antibody fragment disclosed herein.
- the sample may optionally be blood, plasma, lymph, mucus, urine, and/or feces.
- the SARS-CoV S may comprise the amino acid sequence of SEQ ID NO: 401 (SARS-CoV-S, 1288 amino acids, Accession# PDB: 6VSB_B),
- the SARS-CoV-2 may comprise the amino acid sequence of SEQ ID NO: 403 (SARS-CoV -2-S, 1273 amino acids, GenBank: QHD43416.1).
- Such determination methods optionally may comprise an ELISA or radioimmunoassay.
- the subject optionally may be human, a companion animal (e.g., a dog or cat), an agricultural animal, e.g., animals used in meat production, or may comprise an animal in a zoo, e.g. , a tiger or lion.
- a companion animal e.g., a dog or cat
- an agricultural animal e.g., animals used in meat production
- an animal in a zoo e.g. , a tiger or lion.
- the samples optionally may be collected at different times from the subject and the presence or absence or the level of SARS-CoV-S or SARS-CoV-S-2 or another coronavirus S protein homologous thereto may be detected in order to assess whether the subject has recovered.
- the SARS-CoV S may comprise the amino acid sequence of SEQ ID NO: 401 (SARS-CoV-S, 1288 amino acids, Accession# PDB: 6VSB_B), and optionally the SARS-CoV-2 may comprise the amino acid sequence of SEQ ID NO: 403 (SARS-CoV -2-S, 1273 amino acids, GenBank: QHD43416.1).
- the present disclosure further provides methods of inducing an immune response against SARS-CoV or SARS-CoV-2 or another coronavirus, which may be selected from MERS-CoV, HCoV-HKUl, HCoV-OC43, HCoV-229E, and HCoV-NL63, in a subject in need thereof.
- the methods may comprise administering at least one antibody or antigen-binding antibody fragment of the present disclosure.
- the methods may comprise administering a cocktail of different antibodies or antigen-binding antibody fragments of the present disclosure, e.g., which bind to the same or different epitopes on the same or different CoV-Ss.
- the immune response elicits immunoprotection, optionally prolonged, against at least one coronavirus, optionally SARS-CoV or SARS-CoV-2, further optionally against another coronavirus.
- the present disclosure further provides methods of inhibiting or blocking infection of susceptible cells by SARS-CoV or SARS-CoV-2 or another coronavirus, such as MERS-CoV, HCoV- HKU1, HCoV-OC43, HCoV-229E, and HCoV-NL63, in a subject in need thereof.
- SARS-CoV or SARS-CoV-2 or another coronavirus such as MERS-CoV, HCoV- HKU1, HCoV-OC43, HCoV-229E, and HCoV-NL63
- the method may comprise administering at least one antibody or antigen-binding antibody fragment, of the present disclosure, e.g., a cocktail as above-described.
- the present disclosure further provides methods of treating infection by SARS-CoV or SARS-CoV-2 or another coronavirus optionally such as MERS-CoV, HCoV-HKUl, HCoV-OC43, HCoV-229E, and HCoV-NL63, or treating a condition, symptom, disease, or disorder associated with said infection in a subject in need thereof.
- the method may comprise administering to the subject a therapeutically effective amount of at least one antibody or antigen-binding antibody fragment, an ADC or a CAR, of the present disclosure, e.g., a cocktail as above-described.
- the condition, symptom, disease, or disorder comprises at least one of bronchitis, pneumonia, respiratory failure, acute respiratory failure, organ failure, multi-organ system failure, pediatric inflammatory multisystem syndrome, acute respiratory distress syndrome, blood clot, a cardiac condition, myocardial injury, myocarditis, heart failure, cardiac arrest, acute myocardial infarction, dysrhythmia, venous thromboembolism, post-intensive care syndrome, shock, anaphylactic shock, cytokine release syndrome, septic shock, disseminated intravascular coagulation, ischemic stroke, intracerebral hemorrhage, microangiopathic thrombosis, psychosis, seizure, nonconvulsive status epilepticus, traumatic brain injury, stroke, anoxic brain injury, encephalitis, posterior reversible leukoencephalopathy, necrotizing encephalopathy, post-infectious encephalitis, autoimmune mediated encephalitis, acute disse
- the present disclosure also provides methods of preventing infection by SARS-CoV or SARS-CoV-2 or another coronavirus optionally selected from the group consisting of MERS-CoV, HCoV-HKUl, HCoV-OC43, HCoV-229E, and HCoV-NL63 in a subject in need thereof.
- the method may comprise administering to the subject a prophylactically effective amount of at least one antibody or antigen-binding antibody fragment, an ADC or a CAR, of the present disclosure, e.g., a cocktail as above-described.
- the present disclosure also provides methods of preventing the need for a subject infected with SARS-CoV or SARS-CoV-2 or another coronavirus optionally selected from the group consisting of MERS-CoV, HCoV-HKUl, HCoV-OC43, HCoV-229E, and HCoV-NL63 to be placed on a ventilator, or reducing the time that a subject infected with SARS-CoV or SARS-CoV-2 or another coronavirus optionally selected from the group consisting of MERS-CoV, HCoV-HKUl, HCoV-OC43, HCoV-229E, and HCoV-NL63 is on a ventilator.
- the method may comprise administering to the subject a prophylactically or therapeutically effective amount of at least one antibody or antigen-binding antibody fragment, an ADC or a CAR, of the present disclosure, e.g., a cocktail as above-described.
- the present disclosure provides methods of preventing the onset of pneumonia in a subject infected SARS-CoV or SARS-CoV-2 or another coronavirus optionally selected from the group consisting of MERS-CoV, HCoV-HKUl, HCoV-OC43, HCoV-229E, and HCoV-NL63, or treating pneumonia and/or the symptoms of pneumonia in a subject for a subject infected SARS-CoV or SARS-CoV-2 or another coronavirus optionally selected from the group consisting of MERS-CoV, HCoV-HKUl, HCoV-OC43, HCoV-229E, and HCoV-NL63.
- the antibody, or antigen-binding fragment thereof is administered within 5 days of symptom onset. In some embodiments, the antibody, or antigen-binding fragment thereof, is administered within 4 days of symptom onset. In some embodiments, the antibody, or antigen-binding fragment thereof, is administered within 3 days of symptom onset. In some embodiments, the antibody, or antigen-binding fragment thereof, is administered within 2 days of symptom onset. In some embodiments, the antibody, or antigen-binding fragment thereof, is administered within 1 day of symptom onset.
- the method may comprise administering to the subject a prophylactically or therapeutically effective amount of at least one antibody or antigen-binding antibody fragment, an ADC or a CAR, of the present disclosure, e.g., a cocktail as above-described.
- the subject optionally may be human or may comprise a companion animal, agricultural animal or animal in a zoo.
- the subject is an adult. In some embodiment, the subject is an adolescent. In some embodiments, the subject is a child, or a pediatric subject. In one embodiment, the subject is a child, e.g., from birth to age of 11 years old, e.g., birth to ⁇ 1 month, 1 month to ⁇ 2 years, 5-11 years old, or 2-11 years old. In other embodiments, the adolescent is 12-17 years old or 12-15 years old. In one embodiment, the adult is over the age of 18. In some embodiments, the subject has no known recent exposure to COVID-19. In some embodiments, the subject has a known recent exposure to COVID-19.
- recent exposure is within the last 14 days, within the last 10 days, within the last 7 days, within the last 6 days, 5 days, 4 days, 3 days, 2 days, or 24 hours.
- the subject has COVID-19 symptoms or an active COVID-19 infection.
- the subject e.g., the adult, the adolescent, or the child, is vaccinated or has received a COVID-19 vacination.
- the subject e.g., the adult, the adolescent, or the child, is not vaccinated or has not received a COVID-19 vacination.
- the subject is immunocompromised.
- the subject is a significantly immune compromised subject, e.g., adult subject or pediatric subject, but whose underlying medical condition(s) put them at increased risk of developing severe symptomatic COVID-19.
- the subject has no known recent exposure to COVID-19.
- the subject has a known recent exposure to COVID-19.
- recent exposure is within the last 14 days, within the last 10 days, within the last 7 days, within the last 6 days, 5 days, 4 days, 3 days, 2 days, or 24 hours.
- the subject has COVID-19 symptoms or an active COVID-19 infection.
- the immunocompromsed subject includes, but is not limited to, an individual who is actively treated for solid tumor or hematologic malignancies; solid organ transplant (SOT) recipient taking immunosuppressive therapy; CAR-T-cell therapy or HCT recipient; subjects having moderate or severe primary immunodeficiency; subjects having advanced or untreated HIV infection, or who are taking high-dose corticosteroids, alkylating agents, antimetabolites, or TNF blockers.
- the immunocompromised subject is vaccinated or has received a COVID-19 vacination. In some embodiments, the immunocompromised subject is not vaccinated or has not received a COVID-19 vacination.
- the subject is at a high risk of COVID progression.
- the subjects at a high risk of disease progression are age 50 years old or above with no comorbid conditions or additional risk factors for progression of COVID-19.
- the subjects at a high risk of disease progression are those about 18 to about 50 years old and with one or more preexisting medical conditons selected from the group consisting of (i) obesity (body mass index (BMI) > 30 kg/m 2 ), (ii) diabetes (type 1 or type 2); (iii) chronic kidney disease (eGFR calculated by modification of diet in renal disease (MDRD) of 59 mL/min/1.73 m 2 or less, including end-stage renal disease on hemodialysis); (iv) chronic lung disease (emphysema/chronic obstructive pulmonary disease, chronic bronchitis, interstitial lung disease [including idiopathic pulmonary fibrosis], cystic fibrosis, or moderate to severe asthma [defined as requiring daily therapy]); (v) cardiac disease (heart failure, coronary artery disease, cardiomyopathies, or hypertension [with at least one medication prescribed or recommended]); (vi) sickle cell disease or thalassemia
- the subjects at a high risk of disease progression are those about 12 to about 17 years old (inclusive) and with one or more preexisting medical conditions selected from the group consisting of (i) BMI >85th percentile for age and sex based on United States Center for Disease Control (CDC) growth charts; (ii) diabetes (Type 1 or Type 2); (iii) chronic kidney disease; (iv) sickle cell disease or thalassemia; (v) congenital or acquired heart disease; (vi) neurodevelopmental disorders (e.g., cerebral palsy, Down syndrome); (vii) a medically-related technological dependence (e.g., tracheostomy, gastrostomy, or positive pressure ventilation not related to COVID-19); (viii) asthma, reactive airway or other chronic respiratory disease that requires daily medication for control; (ix) solid organ or blood stem cell transplant recipients; (x) other immunodeficiency due to underlying illness or immunosuppressant medication; (xi) substance use disorder; or (x
- the subject is age 50 or above with no comorbid conditions or additional risk factors for progression of COVID-19.
- the subject has hypertension with at least one medication prescribed or recommended.
- the subject has moderate to severe asthma requiring daily therapy.
- the subject may have at least one risk factor which renders them more prone to a poor clinical outcome.
- the risk factors may comprise one or more of (i) advanced age such as over 55, 60 or 65 years old, (ii) diabetes, (iii) a chronic respiratory condition such as asthma, cystic fibrosis, another fibrotic condition, or COPD, (iv) obesity, (iv) hypertension, (v) a cardiac or cardiovascular condition, such as heart defects or abnormalities, (vi) a chronic inflammatory or autoimmune condition, e.g., lupus or multiple sclerosis, and (vii) an immunocompromised status which optionally may be caused by cancer, chemotherapy, smoking, bone marrow or organ transplantation, immune deficiencies, poorly controlled HIV infection or AIDS, or prolonged use of corticosteroids or other immunosuppressive medications.
- the subject may further be treated with at least one other drug.
- the method further comprises administering to the subject at least one other drug.
- such one other drug may be: (i) an antiviral drug, optionally, remdesivir, favipiravir, darunavir, nelfinavir, saquinavir, lopinavir, or ritonavir; (ii) an antihelminth drug, optionally ivermectin; (iii) an antiparasitic drug, optionally hydroxychloroquine, chloroquine, or atovaquone;
- antibacterial vaccine optionally the tuberculosis vaccine BCG
- an anti-inflammatory drug optionally a steroid such as ciclesonide, a TNF inhibitor (e.g., adalimumab), a TNF receptor inhibitor (e.g., etanercept), an IL-6 inhibitor (e.g., clazakizumab), an IL-6 receptor inhibitor (e.g., tocilizumab), or metamizole
- an antihistamine drug optionally bepotastine
- an ACE inhibitor optionally moexipril
- a drug that inhibits priming of CoV-S optionally a serine protease inhibitor, further optionally nafamostat.
- the subject may further be treated with: (I) an antiviral agent, optionally, remdesivir, favipiravir, darunavir, nelfinavir, saquinavir, lopinavir, or ritonavir; and (II) at least one other drug.
- the method may further comprise administering to the subject (I) an antiviral agent, optionally, remdesivir, favipiravir, darunavir, nelfinavir, saquinavir, lopinavir, or ritonavir; and (II) at least one other drug.
- the at least one other drug may be (i) an antihelminth drug, further optionally ivermectin; (ii) an antiparasitic drug, optionally hydroxychloroquine, chloroquine, or atovaquone; (iii) an antibacterial vaccine, which is optionally the tuberculosis vaccine BCG; or (iv) an anti-inflammatory drug, optionally a steroid such as ciclesonide, a TNF inhibitor (e.g., adalimumab), a TNF receptor inhibitor (e.g., etanercept), an IF-6 inhibitor (e.g., clazakizumab), an IF-6 receptor inhibitor (e.g., toclizumab), or metamizole; (v) an antihistamine drug, optionally bepotastine; (vi) an ACE inhibitor, optionally moexipril; and/or (vii) a drug that inhibits priming of CoV-S, which is optionally
- the subject may further be treated with a vaccine, or has been treated with a vaccine, e.g., a COVID-19 vaccine.
- a vaccine e.g., a COVID-19 vaccine.
- the anti-CoV-S antibodies or antigen-binding fragments thereof are administered as a vaccine supplement.
- the term “vaccine supplement” refers to a treatment that is administered prior to, concurrently with, or after, receiving a vaccine treatment, e.g., a COVID-19 vaccine.
- the vaccine supplement may be administered instead of a vaccine booster or an additional dose.
- the anti-CoV-S antibody, or antigen-binding fragment thereof is administered in combination with a vaccine. In some embodiments, the anti-CoV-S antibody, or antigen-binding fragment thereof, is administered concurrently with a COVID-19 vaccine. In some embodiments, the anti-CoV-S antibody, or antigen-binding fragment thereof, is administered after a COVID-19 vaccine. In some embodiments, the anti-CoV-S antibody, or antigen-binding fragment thereof, is administered prior to COVID-19 vaccine.
- the anti-CoV-S antibodies, or antigen -binding fragments thereof are administered concurrently with a COVID-19 vaccine, within 24 hours of a COVID-19 vaccine, within 48 hours of a COVID-19 vaccine, within 3 days, 4 days, 5 days, 6 days, 7 days, 10 days, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months or 12 months of a COVID-19 vaccine.
- the COVID-19 vaccine is an mRNA vaccine.
- the COVID- 19 vaccine is COMIRNATY (by PFIZER, also known as BNT 162b2), the Moderna COVID-19 vaccine (mRNA-1273), the Johnson & Johnson COVID-19 vaccine (JNJ-78436735), or the AstraZeneca COVID-19 vaccine.
- anti-CoV-S antibodies or antigen-binding antibody fragments of the present disclosure may be characterized by having a certain VH CDR3 sequences or having a VH CDR3 sequences that are similar to a certain VH CDR3.
- antibody or antigen-binding antibody fragment of the present disclosure may comprise an Fc region.
- the Fc region may comprise a wild type sequence or a variant sequence and optionally may comprise an amino acid sequence of SEQ ID NOs: 411, 412, 413, 414, 415, 416, or 417.
- the isolated antibody or antigen-binding antibody fragment may bind to the SI subunit of SARS-CoV-S or of SARS-CoV-2-S.
- the isolated antibody or antigen-binding antibody fragment may bind to the receptor binding domain (RBD) or the N-terminal domain (NTD) of SARS-CoV-S or of SARS- CoV-2-S.
- RBD receptor binding domain
- NTD N-terminal domain
- the isolated antibody or antigen-binding antibody fragment may bind to the ACE2-binding motif of SARS-CoV-S or of SARS-CoV-2-S and optionally further binds to the epitope of the antibody CR3022.
- the isolated antibody or antigen-binding antibody fragment may compete with ACE2.
- the isolated antibody or antigen-binding antibody fragment may compete with: (i) ACE2 and the antibody CR3022; or (ii) ACE2 but not the antibody CR3022.
- the isolated antibody or antigen-binding antibody fragment (a) may bind to the S protein of SARS-CoV and/or of SARS-CoV-2; and (b) may not bind to any of the S proteins of HCoV-229E, HCoV-HKUl, HCoV-NL63, and HCoV-OK43.
- the isolated antibody or antigen-binding antibody fragment may (a) bind to the S protein of SARS-CoV and/or of SARS-CoV-2; and also (b) bind to the S protein of at least one of HCoV-229E, HCoV-HKUl, HCoV-NL63, and HCoV-OK43.
- the isolated antibody or antigen-binding antibody fragment may neutralize SARS-CoV and/or SARS-CoV-2.
- the isolated antibody or antigen-binding antibody fragment may neutralize SARS-CoV and/or SARS-CoV-2 at 100 nM in vitro.
- the isolated antibody or antigen-binding antibody fragment may neutralize SARS-CoV and/or SARS-CoV-2 at: (i) an IC50 of about 100 nM or lower, of about 50 nM or lower, of about 20 nM or lower, of about 10 nM or lower, of about 5 nM or lower, of about 2 nM or lower, of about 1 nM or lower, of about 500 pM or lower, of about 200 pM or lower, of about 100 pM or lower, of about 50 pM or lower, of about 20 pM or lower, of about 10 pM or lower, of about 5 pM or lower, of about 2 pM or lower, or of about 1 pM or lower; and/or (ii) an IC50 of about 5 ⁇ g/mL or lower, of about 4.5 ⁇ g/mL or lower, of about 4 ⁇ g/mL or lower, of about 3.5 ⁇ g/mL or lower, of about 3
- the isolated antibody or antigen-binding antibody fragment may neutralize SARS-CoV and/or SARS-CoV-2 at an IC90 of about 10 ⁇ g/mL or less, an IC90 of about 1 to about 10 ⁇ g/mL, an IC90 of about 2 to about 10 ⁇ g/mL, an IC90 of about 3 to about 10 ⁇ g/mL, an IC90 of about 5 to 10 ⁇ g/mL, an IC90 of about 7 to 10 ⁇ g/mL, an IC90 of about 8 to 10 ⁇ g/mL, an IC90 of about 9 to 10 ⁇ g/mL, an IC90 of about 10 ⁇ g/mL, 9 ⁇ g/mL, 8 ⁇ g/mL, 7, ⁇ g/mL, 6 ⁇ g/mL, or 5 ⁇ g/mL.
- the isolated antibody or antigen-binding antibody fragment may bind to CoV-S (S protein of any CoV, such as but not limited to SARS-CoV-S and/or SARS-CoV -2-S) with a KD value of: (i) 100 nM or lower; (ii) 10 nM or lower; (iii) 1 nM or lower; (iv) 100 pM or lower; (v) 10 pM or lower; (vi) 1 pM or lower; or (vii) 0.1 pM or lower.
- CoV-S S protein of any CoV, such as but not limited to SARS-CoV-S and/or SARS-CoV -2-S
- KD value of: (i) 100 nM or lower; (ii) 10 nM or lower; (iii) 1 nM or lower; (iv) 100 pM or lower; (v) 10 pM or lower; (vi) 1 pM or lower; or (vii) 0.1 p
- the antibody, or antigen-binding fragment thereof is administered intravenously. In other embodiments, the antibody, or antigen-binding fragment thereof, is administered intramuscularly.
- the antibody, or antigen-binding fragment thereof is administered. In some embodiments, at least two of the antibody, or antigen-binding fragment thereof, are administered.
- the anti-CoV-S antibody and antigen-binding fragment thereof e.g., ADI-58125
- the anti-CoV-S antibody and antigen-binding fragment thereof may be used in combination with a second antibody, or antigen-binding fragment thereof, wherein the second antibody, or antigen-binding fragment thereof, is selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, ADI-58131, or a combination thereof.
- the second antibody, or antigen-binding fragment thereof is ADI-58122. In one embodiment, the second antibody, or antigen-binding fragment thereof, is ADI-58127. In one embodiment, the second antibody, or antigen-binding fragment thereof, is ADI-58129. In one embodiment, the second antibody, or antigen-binding fragment thereof, is ADI-58131.
- the antibody, or antigen-binding fragment thereof is administered at a dose of about 100 mg to about 5000 mg, about 100 mg to about 4500 mg, about 100 mg to about 4000 mg, about 100 mg to about 3500 mg, about 100 mg to about 3000 mg, about 100 mg to about 2500 mg, about 300 mg to about 4500 mg, about 500 mg to about 4500 mg, about 600 mg to about 4500 mg, about 1200 mg to about 4500 mg, about 100 mg to about 2000 mg, about 200 mg to about 1500 mg, about 300 mg to about 600 mg, about 500 mg to about 1200 mg, or about 300 mg to about 1200 mg.
- the antibody, or antigen-binding fragment thereof is administered at a dose of about 150 mg, about 200 mg, about 300 mg, about 400 mg, about 450 mg, about 500 mg, about 600 mg, about 700 mg, about 800 mg, about 900 mg, about 1000 mg about 1100 mg, about 1200 mg, about 1300 mg, about 1400 mg, about 1500 mg, about 1600 mg, about 1700 mg, about 1800 mg, about 1900 mg, about 2000 mg, about 2500 mg, about 3000 mg, about 3500 mg, about 4000 mg, about 4500 mg, or about 5000 mg.
- the antibody, or antigen-binding fragment thereof is administered at a dose of about 150 mg intramuscularly, about 300 mg intramuscularly, about 450 mg intramuscularly, about 500 mg intravenously, about 600 mg intramuscularly, about 1200 mg intramuscularly, about 1200 mg intravenously, about 4500 mg intramuscularly, or about 4500 mg intravenously.
- the antibody, or antigen-binding fragment thereof is administered at a dosage determined based on the current coronavirus variant(s) that is (are) currently circulating in the population (also referred to herein as “variant-based dosing”).
- the antibody, or antigen-binding fragment thereof is administered intramuscularly as a single 1200 mg dose.
- the antibody, or antigen-binding fragment thereof is administered intravenously as a 1200 mg dose.
- the antibody, or antigen-binding fragment thereof is administered intravenously as a single 600 mg dose.
- the antibody, or antigen-binding fragment thereof is administered intravenously as a single 500 mg dose.
- the antibody, or antigen-binding fragment thereof is administered intramuscularly as two 600 mg doses (for example, a first 600 mg dose in the right thigh or right arm, and a second 600 mg dose in the left thigh or left arm; or a first 600 mg dose in the left thigh or left arm, and a second 600 mg dose in the right thigh or right arm) on the same day, for a total dosage of 1200 mg.
- two 600 mg doses for example, a first 600 mg dose in the right thigh or right arm, and a second 600 mg dose in the left thigh or left arm; or a first 600 mg dose in the left thigh or left arm, and a second 600 mg dose in the right thigh or right arm
- the antibody, or antigen-binding fragment thereof is administered intramuscularly as two 300 mg doses (for example, a first 300 mg dose in the right thigh or right arm, and a second 300 mg dose in the left thigh or left arm; or a first 300 mg dose in the left thigh or left arm, and a second 300 mg dose in the right thigh or right arm) on the same day, for a total dosage of 600 mg.
- two 300 mg doses for example, a first 300 mg dose in the right thigh or right arm, and a second 300 mg dose in the left thigh or left arm; or a first 300 mg dose in the left thigh or left arm, and a second 300 mg dose in the right thigh or right arm
- the antibody, or antigen-binding fragment thereof is administered once. In one embodiment, the antibody, or antigen-binding fragment thereof, is administered weekly. In another embodiment, the antibody, or antigen-binding fragment thereof, is administered daily, weekly, every two weeks, monthly, or every two months. In one embodiment, the antibody, or antigen-binding fragment thereof, is administered weekly for about four weeks, once weekly for about a month, weekly for about 5 weeks, weekly for about 6 weeks, weekly for about 7 weeks, or weekly for about two months.
- the methods further comprise obtaining a serum sample from the subject.
- the antibody, or antigen-binding fragment thereof reaches a maximum concentration (Cmax) of about 10 ⁇ g/mL to about 1000 ⁇ g/mL, about 20 ⁇ g/mL to about 500 ⁇ g/mL, about 30 ⁇ g/mL to about 400 ⁇ g/mL, about 40 ⁇ g/mL to about 300 ⁇ g/mL, about 50 ⁇ g/mL to about 200 ⁇ g/mL, about 30 ⁇ g/mL to about 200 ⁇ g/mL, about 100 ⁇ g/mL to about 200 ⁇ g/mL, about 50 ⁇ g/mL to about 100 ⁇ g/mL, or about 30 ⁇ g/mL to about 70 ⁇ g/mL, about 100 ⁇ g/mL to about 400 ⁇ g/mL, or about 150 ⁇ g/mL to about 350 ⁇ g/mL in the
- the antibody, or antigen-binding fragment thereof reaches a maximum concentration (Cmax) of about 30 ⁇ g/mL, about 40 ⁇ g/mL, about 50 ⁇ g/mL, about 60 ⁇ g/mL, about 70 ⁇ g/mL, about 80 ⁇ g/mL, about 90 ⁇ g/mL, about 100 ⁇ g/mL, about 110 ⁇ g/mL, about 120 ⁇ g/mL, about 130 ⁇ g/mL, about 140 ⁇ g/mL, about 150 ⁇ g/mL, about 160 ⁇ g/mL, about 170 ⁇ g/mL, about 180 ⁇ g/mL, about 190 ⁇ g/mL, or about 200 ⁇ g/mL, about 210 ⁇ g/mL, about 220 ⁇ g/mL, about 230 ⁇ g/mL, about 240 ⁇ g/mL, about 250 ⁇ g/mL, about 260 ⁇ g/mL, about 270
- the median time for the antibody, or antigen-binding fragment thereof, to reach a maximum concentration in the serum sample of the subject is about 0.01-30 days, about 0.01-0.05 days, about 5-30 days, about 3-20 days, about 6-20 days, about 7-18 days, or about 8-15 days, or about 13-15 days after administration. In some embodiments, the median time for the antibody, or antigen-binding fragment thereof, to reach a maximum concentration in the serum sample of the subject is about 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 days after administration.
- the median time for the antibody, or antigen-binding fragment thereof, to reach a maximum concentration in the serum sample of the subject is less than about 1 day, e.g., about 0.01, about 0.02, about 0.03, about 0.04, about 0.05, about 0.06, about 0.07, about 0.08, about 0.09, about 0.1, about 0.2, about 0.3, about 0.4, about 0.5, about 0.6, about 0.7, about 0.8, or about 0.9 days after administration.
- the antibody, or antigen-binding fragment thereof reaches a maximum concentration (Cmax) of about 30 ⁇ g/mL to about 200 ⁇ g/mL, about 100 ⁇ g/mL to about 200 ⁇ g/mL, about 30 ⁇ g/mL to about 100 ⁇ g/mL, about 40 ⁇ g/mL to about 80 ⁇ g/mL, about 50 ⁇ g/mL to about 70 ⁇ g/mL, or about 30 ⁇ g/mL to about 65 ⁇ g/mL in the serum sample of the subject in about about 0.01-20 days, about 3-20 days, about 6-20 days, about 7-18 days, about 8-15 or about 13-15 days after administration.
- Cmax maximum concentration
- the area under the serum concentration-time curve from day 0 to day 21 is about 100-2000 day* mg/mL, about 1000-2000 day* mg/mL, about 1400-2000 day* mg/mL, about 200-1500 day* mg/mL, about 400-1400 day* mg/mL, about 500-1300 day* mg/mL, about 600-1000 day* mg/mL, or about 800-900 day* mg/mL.
- the area under the serum concentration-time curve from day 0 to day 90 is about 1000-10000 day* mg/mL, about 2000-8000 day* mg/mL, about 2000-5000 day* mg/mL, about 3000-4000 day* mg/mL, about 5000-10000 day* mg/mL, about 2000-4000 day* mg/mL, about 5000-8000 day* mg/mL, or about 6000-8000 day* mg/mL.
- the area under the serum concentration-time curve from day 0 to day 180 is about 1000-10000 day* mg/mL, about 2000-5000 day* mg/mL, about 3000-6000 day* mg/mL, about 5000-10000 day* mg/mL, about 5000-8000 day* mg/mL, or about 4000-6000 day* mg/mL.
- the area under the serum concentration-time curve from day 0 to day 365 is about 1000-30000 day* mg/mL, about 5000-30000 day* mg/mL, about 10000- 30000 day* mg/mL, about 10000-15000 day* mg/mL, about 20000-30000 day* mg/mL, or about 25000-30000 day* mg/mL.
- the antibody, or antigen-binding fragment thereof has a virus neutralizing titer of about 100-2000, about 200-1500, about 300-1500, or about 500-1500 in the serum sample of the subject about 6 months after administration. In some embodiments, the antibody, or antigen-binding fragment thereof, has a virus neutralizing titer of about 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, 1100, 1200, 1300, 1400, 1500, 1600, 1700, 1800, 1900 or 2000 in the serum sample of the subject about 6 months after administration.
- the antibody, or antigen-binding fragment thereof has a serum half- life of about 50-200 days, about 50-160 days, about 50-140 days, about 40-130 days, about 50-100 days, about 60-100 days, about 60-90 days, about 70-90 days, about 90-100 days, about 70-80 days, about 80-130 days, about 100-150 days, about 110-140 days, or about 120-140 days.
- the method further comprises obtaining an epithelial lining fluid (ELF) sample from the subject.
- ELF sample comprises an ELF sample from an upper airway, an lower airway, and/or an alveolar tissue.
- the antibody, or antigen-binding fragment thereof reaches a concentrationof about 1 ⁇ g/mL to about 100 ⁇ g/mL, about 1 ⁇ g/mL to about 80 ⁇ g/mL, about 80 ⁇ g/mL to about 100 ⁇ g/mL, about 50 ⁇ g/mL to about 100 ⁇ g/mL, about 1 ⁇ g/mL to about 50 ⁇ g/mL, about 2 ⁇ g/mL to about 25 ⁇ g/mL, or about 2 ⁇ g/mL to about 10 ⁇ g/mL in the ELF sample of the subject.
- the antibody, or antigen-binding fragment thereof has a receptor occupancy of at least 50%, 60%, 70%, 80%, or 90% in the ELF sample. In some embodiments, the antibody, or antigen-binding ragment thereof, has a receptor occupancy of at least 50%, 60%, 70%, 80%, or 90% in the ELF sample for about 28 days after administration.
- the 80% virus neutralization titer (MN80) of the antibody, or antigen binding fragment thereof, at about days 0-14, about days 7-21, about 3 months, or about 6 months after administration is about 10-6000, about 50-600, about 500-1500, about 1000-2500, about 100- 2500, about 500-2000, about 500-1500, about 400-1200, about 200-1500, about 300-1000, about 400- 800, about 400-1000, or about 500-600 in the subject.
- the 80% virus neutralization titer (MN80) of the antibody, or antigen binding fragment thereof, at about day 7, day 14, or day 21 after administration is about 300-2000, about 400-600, about 600-1500, about 1100-1700, about 500-1700, about 500-1500, about 400-1200, about 400-800, about 400-1000, or about 500-600 in the serum sample of the subject.
- the 80% virus neutralization titer (MN80) of the antibody, or antigen binding fragment thereof, at about 3 months after administration is about 200-1000, about 200-800, about 200-500, about 400-900, or about 400-600 in the serum sample of the subject.
- the 80% virus neutralization titer (MN80) of the antibody, or antigen binding fragment thereof, at about 6 months after administration is about 10-500, about 300-500, or about 50-200 in the serum sample of the subject.
- the 50% virus neutralization titer (MN50) of the antibody, or antigen binding fragment thereof, at about days 0-14, about days 7-21, about 3 months, about 6 months, or about 12 months after administration is about 100-6000, about 300-1500, about 1700-3800, about 3800-5200, about 300-5500, about 1200-4500, about 1300-4300, about 1200-4000, about 100-2500, about 500-2500, about 800-2000, about 1000-1800, about 800-1300, about 900-1100, or about 1300- 1500 in the serum sample of the subject.
- the 50% virus neutralization titer (MN50) of the antibody, or antigen binding fragment thereof, at about day 7, day 14 or day 21 after administration is about 1000-4500, about 1200-4500, about 1300-4300, about 1200-3900, about 1500-4000, about 1800-3800, about 3800-4500, about 1000-1800 or about 1200-1500 in the serum sample of the subject.
- the 50% virus neutralization titer (MN50) of the antibody, or antigen binding fragment thereof, at about 3 months after administration is about 800-1300 or about 900- 1100 in the serum sample of the subject.
- the 50% virus neutralization titer (MN50) of the antibody, or antigen binding fragment thereof, at about 6 months after administration is about 200-500 or about 300-600 in the serum sample of the subject.
- the 50% virus neutralization titer (MN50) of the antibody, or antigen binding fragment thereof, at about 12 months after administration is about 150-500 or about 200-400 in the serum sample of the subject.
- the neutralization titer is determined using a plaque reduction neutralization test (PRNT).
- the serum concentration of the antibody, or antigen-binding fragment thereof is about 1-300 mg/L, about 1-250 mg/L, about 1-200 mg/L, about 1-100 mg/L, about 100-250 mg/L, about 150-200 mg/L, about 120-170 mg/L, about 1-90 mg/L, about 1-80 mg/L, about 1-70 mg/L, about 1-60 mg/L, about 1-50 mg/L, about 1-40 mg/L, about 1-30 mg/L, about 1-20 mg/L, about 1-10 mg/L, about 2-9 mg/L, about 3-8 mg/L, about 4-6 mg/L, about 5-80 mg/L, about 10-70 mg/L, or about 10-60 mg/L in the subject about 3 months after administration.
- the serum concentration of the antibody, or antigen-binding fragment thereof is about 1 mg/L, about 2 mg/L, about 3 mg/L, about 4 mg/L, about 5 mg/L, about 6 mg/L, about 7 mg/L, about 8 mg/L, about 9 mg/L, about 10 mg/L, about 11 mg/L, about 12 mg/L, about 13 mg/L, about 14 mg/L, about 15 mg/L, about 16 mg/L, about 17 mg/L, about 18 mg/L, about 19 mg/L, about 20 mg/L, about 25 mg/L, about 30 mg/L, about 35 mg/L, about 40 mg/L, about 45 mg/L, about 50 mg/L, about 55 mg/L, about 60 mg/L, about 65 mg/L, about 70 mg/L, about 75 mg/L, about 80 mg/L, about 85 mg/L, about 90 mg/L, about 95 mg/L, about 100 mg/L, about 110 mg/L, about 120 mg/L, about
- the serum concentration of the antibody, or antigen-binding fragment thereof is about 1-100 mg/L, about 1-90 mg/L, about 1-80 mg/L, about 1-70 mg/L, about 1-60 mg/L, about 1-50 mg/L, about 1-40 mg/L, about 1-30 mg/L, about 1-20 mg/L, about 1-10 mg/L, about 2-9 mg/L, about 3-8 mg/L, about 4-6 mg/L, about 5-80 mg/L, about 10-70 mg/L, about 10-60 mg/L, about 5-25 mg/L, about 10-30 mg/L, about 15-25 mg/L, about 20-100 mg/L, about 30-100 mg/L, about 50- 80 mg/L, or about 60-70 mg/L in the subject about 6 months after administration.
- the serum concentration of the antibody, or antigen-binding fragment thereof is about 1 mg/L, about 2 mg/L, about 3 mg/L, about 4 mg/L, about 5 mg/L, about 6 mg/L, about 7 mg/L, about 8 mg/L, about 9 mg/L, about 10 mg/L, about 11 mg/L, about 12 mg/L, about 13 mg/L, about 14 mg/L, about 15 mg/L, about 16 mg/L, about 17 mg/L, about 18 mg/L, about 19 mg/L, about 20 mg/L, about 25 mg/L, about 30 mg/L, about 35 mg/L, about 40 mg/L, about 45 mg/L, about 50 mg/L, about 55 mg/L, about 60 mg/L, about 65 mg/L, about 70 mg/L, about 75 mg/L, about 80 mg/L, about 85 mg/L, about 90 mg/L, about 95 mg/L, or about 100 mg/L in the subject about 6 months after administration.
- the serum concentration of the antibody, or antigen-binding fragment thereof is about 0.1-30 mg/L, about 1-30 mg/L, about 1-20 mg/L, about 1-10 mg/L, about 10-20 mg/L, about 0.1-3 mg/L, about 0.5-8 mg/L, about 0.5-10 mg/L, about 2-9 mg/L, about 5-15 mg/L, about 3-8 mg/L, or about 4-6 mg/L in the subject about 12 months after administration.
- the serum concentration of the antibody, or antigen-binding fragment thereof is about 0.1 mg/L, about 0.2 mg/L, about 0.3 mg/L, about 0.4 mg/L, about 0.5 mg/L, about 0.6 mg/L, about 0.7 mg/L, about 0.8 mg/L, about 0.9 mg/L, about 1 mg/L, about 2 mg/L, about 3 mg/L, about 4 mg/L, about 5 mg/L, about 6 mg/L, about 7 mg/L, about 8 mg/L, about 9 mg/L, about 10 mg/L, about 15 mg/L, about 20 mg/L, about 25 mg/L, or about 30 mg/L in the subject about 12 months after administration.
- the clearance rate for the antibody, or antigen-binding fragment thereof is about 0.1-10 mL/d, about 0.1-5.0 mL/d, about 0.1-3.0 mL/d, about 0.5-2.5 mL/d, or about 1.0-2.0 mL/d.
- the steady state volume of distribution for the antibody, or antigen binding fragment thereof is about 1-10 L, about 2-8 L, about 4-9 L, about 4-8 L, or about 5-7L.
- administration of the antibody, or antigen-binding fragment thereof reduces pulmonary inflammation in the subject.
- administration of the antibody, or antigen-binding fragment thereof reduces the risk of COVID-19 hospitalization or death of the subject.
- the risk of COVID-19 hospitalization or death is reduced by at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90% or 95%.
- administration of the antibody, or antigen-binding fragment thereof reduces viral load in the subject.
- the viral load is reduced by at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90% or 95%.
- the viral load is reduced by at least about 0.5 loglO copies/mL, at least about 0.6 loglO copies/mL, at least about 0.7 loglO copies/mL, at least about 0.8 loglO copies/mL, at least about 0.9 loglO copies/mL, at least about 1.0 loglO copies/mL, at least about 1.1 loglO copies/mL, at least about 1.2 loglO copies/mL, at least about 1.3 loglO copies/mL, at least about 1.4 loglO copies/mL, at least about 1.5 loglO copies/mL, at least about 1.6 loglO copies/mL, at least about 1.7 loglO copies/mL, at least about 1.8 loglO copies/mL, at least about 1.9 loglO copies/mL, at least about 2.0 loglO copies/mL, at least about 2.1 loglO copies/mL, at least about 2.2 loglO copies/mL, at least about 2.3 loglO copies/mL
- kits comprising: (a) at least one isolated antibody or antigen-binding antibody fragment disclosed herein; and (b) an instruction for using the antibody or antigen-binding antibody fragment.
- the kit may be for use in: (i) determining whether a CoV is present in a subject; (ii) diagnosing whether a subject has CoV infection; (iii) predicting whether a CoV vaccine will elicit a protective immune response; or (iv) predicting whether a CoV vaccine will elicit a neutralizing antibody response.
- the present disclosure provides a method of inducing an immune response against a coronavirus (CoV) in a subject in need thereof, the method comprising administering to the subject an isolated antibody, or antigen-binding fragment thereof, which binds to the spike protein of a coronavirus (“CoV-S”), wherein said antibody, or antigen-binding fragment thereof, comprises a heavy chain variable region (VH) comprising a VH CDR1 comprising SEQ ID NO:52, a VH CDR2 comprising SEQ ID NO:54, and a VH CDR3 comprising SEQ ID NO:56, and a light chain variable region (VL) comprising a VL CDR1 comprising SEQ ID NO:252, a VL CDR2 comprising SEQ ID NO:254, and a VL CDR3 comprising SEQ ID NO:256; (a) wherein the antibody, or antigen-binding fragment thereof, reaches a maximum concentration (Cmax) of about 10 ⁇
- the present disclosure provides a method of preventing a coronavirus infection in a subject in need thereof, the method comprising administering to the subject an isolated antibody, or antigen-binding fragment thereof, which binds to the spike protein of a coronavirus (“CoV-S”), wherein said antibody, or antigen-binding fragment thereof, comprises a heavy chain variable region (VH) comprising a VH CDR1 comprising SEQ ID NO:52, a VH CDR2 comprising SEQ ID NO:54, and a VH CDR3 comprising SEQ ID NO: 56, and a light chain variable region (VL) comprising a VL CDR1 comprising SEQ ID NO:252, a VL CDR2 comprising SEQ ID NO:254, and a VL CDR3 comprising SEQ ID NO:256; (a) wherein the antibody, or antigen-binding fragment thereof, reaches a maximum concentration (Cmax) of about 10 ⁇ g/mL to about 1000
- the present disclosure provides a method of beating a coronavirus infection in a subject in need thereof, the method comprising administering to the subject an isolated antibody, or antigen-binding fragment thereof, which binds to the spike protein of a coronavirus (“CoV-S”), wherein said antibody, or antigen-binding fragment thereof, comprises a heavy chain variable region (VH) comprising a VH CDR1 comprising SEQ ID NO:52, a VH CDR2 comprising SEQ ID NO:54, and a VH CDR3 comprising SEQ ID NO: 56, and a light chain variable region (VL) comprising a VL CDR1 comprising SEQ ID NO:252, a VL CDR2 comprising SEQ ID NO:254, and a VL CDR3 comprising SEQ ID NO:256; (a) wherein the antibody, or antigen-binding fragment thereof, reaches a maximum concentration (Cmax) of about 10 ⁇ g/mL to about 1000 ⁇
- Cmax maximum concentration
- the present disclosure provides a method of treating a symptom of a coronavirus infection in a subject in need thereof, the method comprising administering to the subject an isolated antibody, or antigen-binding fragment thereof, which binds to the spike protein of a coronavirus (“CoV-S”), wherein said antibody, or antigen-binding fragment thereof, comprises a heavy chain variable region (VH) comprising a VH CDR1 comprising SEQ ID NO:52, a VH CDR2 comprising SEQ ID NO:54, and a VH CDR3 comprising SEQ ID NO:56, and a light chain variable region (VL) comprising a VL CDR1 comprising SEQ ID NO:252, a VL CDR2 comprising SEQ ID NO:254, and a VL CDR3 comprising SEQ ID NO:256; (a) wherein the antibody, or antigen-binding fragment thereof, reaches a maximum concentration (Cmax) of about 10 ⁇ g/mL
- VH heavy chain variable
- the symptom comprises at least one of bronchitis, pneumonia, respiratory failure, acute respiratory failure, organ failure, multi-organ system failure, pediatric inflammatory multisystem syndrome, acute respiratory distress syndrome, blood clot, a cardiac condition, myocardial injury, myocarditis, heart failure, cardiac arrest, acute myocardial infarction, dysrhythmia, venous thromboembolism, post-intensive care syndrome, shock, anaphylactic shock, cytokine release syndrome, septic shock, disseminated intravascular coagulation, ischemic stroke, intracerebral hemorrhage, microangiopathic thrombosis, psychosis, seizure, nonconvulsive status epilepticus, traumatic brain injury, stroke, anoxic brain injury, encephalitis, posterior reversible leukoencephalopathy, necrotizing encephalopathy, post-infectious encephalitis, autoimmune mediated encephalitis, acute disseminated encephalomy
- the present disclosure provides a method of decreasing the risk of mortality, hospitalization, mechanical ventilation, or a combination thereof, in a subject infected by a coronavirus, the method comprising administering to the subject an isolated antibody, or antigen binding fragment thereof, which binds to the spike protein of a coronavirus (“CoV-S”), wherein said antibody, or antigen-binding fragment thereof, comprises a heavy chain variable region (VH) comprising a VH CDR1 comprising SEQ ID NO:52, a VH CDR2 comprising SEQ ID NO:54, and a VH CDR3 comprising SEQ ID NO: 56, and a light chain variable region (VL) comprising a VL CDR1 comprising SEQ ID NO:252, a VL CDR2 comprising SEQ ID NO:254, and a VL CDR3 comprising SEQ ID NO:256; (a) wherein the antibody, or antigen-binding fragment thereof, reaches a maximum concentration (Cmax)
- the CoV-S is the spike protein of SARS-CoV (“SARS-CoV-S”) or the spike protein of SARS-CoV-2 (“SARS-CoV-2-S”).
- the antibody, or antigen-binding fragment thereof cross-reacts with SARS-CoV-S and SARS-CoV-2-S.
- SARS-CoV-S comprises an amino acid sequence of SEQ ID NO: 401, and wherein SARS-CoV-2-S comprises an amino acid sequence of SEQ ID NO: 403.
- the SARS-CoV -2-S is a B.1.1.7 variant, a B. 1.351 variant, a B.1.1.28 variant, a B. 1.429 variant, a P.l variant, a B.1.617 variant, a B.1.617.2 variant, a C.37 variant, a 1.621 variant, a AY.l variant, a 1.623 variant, a C.36 variant, a A.27 variant, a AV.l variant, a B.1.1.482 variant, a B.1.1.523 variant, a B.1.427 variant, a AY.4 variant, a AY.ll variant, a D614G variant of SEQ ID NO: 403, a B.1.1.529/BA.1 variant, a BA.1.1 variant, or a BA.2 variant.
- the present disclosure provides a method for inducing an immune response against a coronavirus Omicron variant in a subject in need thereof, for treating a coronavirus infection caused by an Omicron variant in a subject in need thereof, for preventing a coronavirus infection caused by an Omicron variant in a subject in need thereof, for treating a symptom of an infection of a subject by an Omicron variant in a subject in need thereof, and/or for decreasing the risk of mortality, hospitalization, mechanical ventilation, or a combination thereof, in a subject infected by an Omicron variant, the method comprising administering to the subject an isolated antibody, or antigen-binding fragment thereof, wherein said antibody, or antigen-binding fragment thereof, comprises a heavy chain variable region (VH) comprising a VH CDR1 comprising SEQ ID NO:52, a VH CDR2 comprising SEQ ID NO:54, and a VH CDR3 comprising SEQ ID NO:56, and a light
- VH heavy chain variable
- the Omicron variant comprises a B.1.1.529/BA.1 variant, a BA.1.1 variant, or a BA.2 variant.
- the VH comprises a sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to SEQ ID NO:58 and the VL comprises a sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to SEQ ID NO:258.
- the VH comprises or consists of SEQ ID NO:58 and the VL comprises or consists of SEQ ID NO:258.
- the method may further comprise selecting a subject.
- the method may further comprise selecting a subject that would benefit from administration of the antibody, or antigen-binding fragment thereof.
- the subject is a human subject. In some embodiments, the subject is an adult, an adolescent, or a child.
- the subject has at least one risk factor which renders them more prone to a poor clinical outcome.
- the at least one risk factor is selected from the group consisting of: an old age selected from the group consisting of over 55, over 60 or over 65 years old; diabetes, a chronic respiratory condition, obesity, hypertension, a cardiac or cardiovascular condition, a chronic inflammatory or autoimmune condition, and an immune compromised status.
- the subject is immunocompromised. In some embodiments, the subject is at a high risk of disease progression.
- the subject is (a) age 50 years or above with no comorbid conditions or additional risk factors for progression of COVID-19; (b) between about 18 to about 50 years old and with one or more preexisting medical conditions selected from the group consisting of obesity, diabetes, chronic kidney disease, chronic lung disease, cardiac disease, sickle cell disease or thalassemia, solid organ or blood stem cell transplant recipient, other immunodeficiency due to underlying illness or immunosuppressant medication, Down Syndrome, stroke or cerebrovascular disease, substance use disorder, and pregnancy; or (c) between about 12 to about 17 years old and with one or more preexisting medical conditions selected from the group consisting of obesity, diabetes, chronic kidney disease, sickle cell disease or thalassemia, congenital or acquired heart disease, neurodevelopmental disorder, a medically-related technological dependence, asthma or chornic respiratory disease, solid organ or blood stem cell transplant recipient, other immunodeficiency due to underlying illness or immunosuppressant medication, Down Syndrome, stroke or cerebrovascular disease, substance use disorder, and
- the subject is age 50 or above with no comorbid conditions or additional risk factors for progression of COVID-19.
- the subject has hypertension with at least one medication prescribed or recommended.
- the subject has moderate to severe asthma requiring daily therapy.
- the antibody, or the antigen-binding fragment thereof is administered intramuscularly or intravenously.
- the antibody, or the antigen-binding fragment thereof is administered at a dose of about 100 mg to about 5000 mg, about 300 mg to about 4500 mg, about 500 mg to about 4500 mg, about 1200 mg to about 4500 mg, about 100 mg to about 2000 mg, about 200 mg to about 1500 mg, about 300 mg to about 600 mg, about 500 mg to about 1200 mg, about 300 mg to about 1200 mg; or at a dose of about 4500 mg, a dose of about 1200 mg, a dose of about 600 mg, or a dose of about 500 mg; or as two doses of 600 mg each on the same day, or as two doses of 300 mg each on the same day.
- the antibody, or antigen-binding fragment thereof is administered once, or is administered weekly.
- the method further comprises obtaining an epithelial lining fluid (ELF) sample from the subject.
- ELF epithelial lining fluid
- the ELF sample comprises an ELF sample from an upper airway, an lower airway, and/or an alveolar tissue.
- the antibody, or antigen-binding fragment thereof reaches a concentration of about 1 ⁇ g/mL to about 100 ⁇ g/mL, about 1 ⁇ g/mL to about 80 ⁇ g/mL, about 80 ⁇ g/mL to about 100 ⁇ g/mL, about 50 ⁇ g/mL to about 100 ⁇ g/mL, about 1 ⁇ g/mL to about 50 ⁇ g/mL, about 2 ⁇ g/mL to about 25 ⁇ g/mL, or about 2 ⁇ g/mL to about 10 ⁇ g/mL, in the ELF sample of the subject.
- the antibody, or antigen-binding fragment thereof has a receptor occupancy of at least 50%, 60%, 70%, 80%, or 90% in the ELF sample.
- the antibody, or antigen-binding fragment thereof has a receptor occupancy of at least 50%, 60%, 70%, 80%, or 90% in the ELF sample for about 28 days after administration.
- the antibody, or antigen-binding fragment thereof has a virus neutralizing titer of about 100-2000, about 200-1500, about 300-1500, or about 500-1500 in serum of the subject about 6 months after administration.
- the 80% virus neutralization titer (MN80) of the antibody, or antigen binding fragment thereof, at about days 0-14, about days 7-21, about 3 months, or about 6 months after administration is about 10-6000, about 50-600, about 500-1500, about 1000-2500, about 100- 2500, about 500-2000, about 500-1500, about 400-1200, about 200-1500, about 300-1000, about 400- 800, about 400-1000, or about 500-600 in the subject.
- the 80% virus neutralization titer (MN80) of the antibody, or antigen binding fragment thereof, at about day 7, day 14, or day 21 after administration is about 300-2000, about 400-600, about 600-1500, about 1100-1700, about 500-1700, about 500-1500, about 400-1200, about 400-800, about 400-1000, or about 500-600 in the serum sample of the subject.
- the 80% virus neutralization titer (MN80) of the antibody, or antigen binding fragment thereof, at about 3 months after administration is about 200-1000, about 200-800, about 200-500, about 400-900, or about 400-600 in the serum sample of the subject.
- the 80% virus neutralization titer (MN80) of the antibody, or antigen binding fragment thereof, at about 6 months after administration is about 10-500, about 300-500, or about 50-200 in the serum sample of the subject.
- the 50% virus neutralization titer (MN50) of the antibody, or antigen binding fragment thereof, at about days 0-14, about days 7-21, about 3 months, about 6 months, or about 12 months after administration is about 100-6000, about 300-1500, about 1700-3800, about 3800-5200, about 300-5500, about 1200-4500, about 1300-4300, about 1200-4000, about 100-2500, about 500-2500, about 800-2000, about 1000-1800, about 800-1300, about 900-1100, or about 1300- 1500 in the serum sample of the subject.
- the 50% virus neutralization titer (MN50) of the antibody, or antigen binding fragment thereof, at about day 7, day 14 or day 21 after administration is about 1000-4500, about 1200-4500, about 1300-4300, about 1200-3900, about 1500-4000, about 1800-3800, about 3800-4500, about 1000-1800 or about 1200-1500 in the serum sample of the subject.
- the 50% virus neutralization titer (MN50) of the antibody, or antigen binding fragment thereof, at about 3 months after administration is about 800-1300, or about 900- 1100 in the serum sample of the subject.
- the 50% virus neutralization titer (MN50) of the antibody, or antigen binding fragment thereof, at about 6 months after administration is about 200-500, or about 300-600 in the serum sample of the subject.
- the 50% virus neutralization titer (MN50) of the antibody, or antigen binding fragment thereof, at about 12 months after administration is about 150-500, or about 200-400 in the serum sample of the subject.
- the virus neutralization titer is determined using a plaque reduction neutralization test (PRNT).
- PRNT plaque reduction neutralization test
- the serum concentration of the antibody, or antigen-binding fragment thereof is about 1-300 mg/L, about 1-250 mg/L, about 1-200 mg/L, about 1-100 mg/L, about 100-250 mg/L, about 150-200 mg/L, about 120-170 mg/L, about 1-90 mg/L, about 1-80 mg/L, about 1-70 mg/L, about 1-60 mg/L, about 1-50 mg/L, about 1-40 mg/L, about 1-30 mg/L, about 1-20 mg/L, about 1-10 mg/L, about 2-9 mg/L, about 3-8 mg/L, about 4-6 mg/L, about 5-80 mg/L, about 10-70 mg/L, or about 10-60 mg/L in the subject about 3 months after administration.
- the serum concentration of the antibody, or antigen-binding fragment thereof is about 1-100 mg/L, about 1-90 mg/L, about 1-80 mg/L, about 1-70 mg/L, about 1-60 mg/L, about 1-50 mg/L, about 1-40 mg/L, about 1-30 mg/L, about 1-20 mg/L, about 1-10 mg/L, about 2-9 mg/L, about 3-8 mg/L, about 4-6 mg/L, about 5-80 mg/L, about 10-70 mg/L, about 10-60 mg/L, about 5-25 mg/L, about 10-30 mg/L, about 15-25 mg/L, about 20-100 mg/L, about 30-100 mg/L, about 50- 80 mg/L, or about 60-70 mg/L in the subject about 6 months after administration.
- the serum concentration of the antibody, or antigen-binding fragment thereof is about 0.1-30 mg/L, about 1-30 mg/L, about 1-20 mg/L, about 1-10 mg/L, about 10-20 mg/L, about 0.1-3 mg/L, about 0.5-8 mg/L, about 0.5-10 mg/L, about 2-9 mg/L, about 5-15 mg/L, about 3-8 mg/L, or about 4-6 mg/L in the subject about 12 months after administration.
- the clearance rate for the antibody, or antigen-binding fragment thereof is about 0.1-10 mL/d, about 0.1-5.0 mL/d, about 0.1-3.0 mL/d, about 0.5-2.5 mL/d, or about 1.0-2.0 mL/d.
- the steady state volume of distribution for the antibody, or antigen binding fragment thereof is about 1-10 L, about 2-8 L, about 4-9 L, about 4-8 L, or about 5-7L.
- the antibody, or antigen-binding fragment thereof is administered in combination with at least one antibody selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130 and ADI-58131.
- the antibody, or antigen-binding fragment thereof is administered in combination with a vaccine.
- the antibody, or antigen-binding fragment thereof is administered (a) concurrently with a COVID-19 vaccine; (b) after administration of a COVID-19 vaccine, or (c) prior to administration of a COVID-19 vaccine.
- administration of the antibody, or antigen-binding fragment thereof reduces pulmonary inflammation in the subject.
- administration of the antibody, or antigen-binding fragment thereof reduces the risk of COVID-19 hospitalization or death of the subject.
- administration of the antibody, or antigen-binding fragment thereof reduces viral load in the subject.
- methods of predicting the in vivo efficacy of an anti-CoV-S antibody or antigen-binding antibody fragment in preventing or treating CoV infection are provided herein.
- the method may comprise: (a) providing at least one first test subject and at least one second subject or a cell sample derived from at least one first test subject and at least one second subject; (b) administering the antibody or antigen-binding antibody fragment to said at least one first test subject and said at least one second subject or contacting a cell sample from said first and second subject with the antibody or antigen-binding antibody fragment; (c) infecting said at least one first test subject and said at least one second subject with CoV or pseudo CoV or a cell sample obtained from said at least one first test subject and said at least one second subject with CoV or pseudo CoV ; (d) determining whether administration of the antibody or antigen-binding antibody fragment in (b) results in one or more of the following compared to a suitable control: (I) reduction in a CoV-associated symptom; (II) reduction in the CoV viremia; (III) increase in the survival; (IV) increase in the body weight; or (V) reduced infection of cells or virus
- the method may comprise: (a) providing at least one first cell sample and at least one second cell sample; (b) contacting the at least one first cell sample with the antibody or antigen-binding antibody fragment; (c) infecting said at least one first cell sample and at least one second cell sample with CoV or pseudo CoV ; (d) determining whether the antibody or antigen binding antibody fragment results in one or more of the following compared to a suitable control: (I) increase in the cell survival; (II) reduced infection of cells; (III) reduced virus proliferation; (IV) reduced cell stress or death markers; or (V) reduced inflammatory cytokines, in cells of the tested cell sample compared to a control cell sample not contacted with the antibody or antigen-binding antibody fragment.
- the method may comprise: (a) providing at least one first test subject and at least one second subject or a cell sample derived from at least one first test subject and at least one second subject; (b) infecting said at least one first test subject and said at least one second subject with CoV or pseudo CoV or a cell sample derived from at least one first test subject and at least one second subject; (c) administering the antibody or antigen-binding antibody fragment to said at least one second subject or contacting a cell sample derived from at least one first test subject and at least one second subject with the antibody or antigen-binding antibody fragment; (d) determining whether administration of the antibody or antigen-binding antibody fragment in (c) results in one or more of the following: (I) reduction in a CoV-associated symptom; (II) reduction in the CoV viremia; (III) increase in the survival;(IV) increase in the body weight; or (V) reduced infection of cells or virus proliferation in cells in the tested cell sample compared to a control cell
- the method may comprise: (a) providing at least one first cell sample and at least one second cell sample; (b) infecting said at least one first cell sample and at least one second cell sample with CoV or pseudo CoV ; (c) contacting the at least one first cell sample with the antibody or antigen-binding antibody fragment; (d) determining whether the antibody or antigen binding antibody fragment results in one or more of the following compared to a suitable control: (I) increase in the cell survival; (II) reduced infection of cells; (III) reduced virus proliferation; (IV) reduced cell stress or death markers; or (V) reduced inflammatory cytokines, in cells of the tested cell sample compared to a control cell sample not contacted with the antibody or antigen-binding antibody fragment.
- methods of screening for an antibody or antigen-binding antibody fragment that binds to a CoV or CoV-S comprising whether an antibody or antigen-binding antibody fragment comprising 1, 2, 3, 4, 5, or 6 CDRs of any of the antibodies disclosed herein may comprise one or more of the following features: (i) binds to the S protein of a CoV; (ii) binds to the SI subunit of CoV-S; (iii) binds to the RBD of CoV-S; (iv) binds to the NTD of CoV-S; (v) binds to the ACE2-binding motif of CoV-S; (vi) competes with ACE2; (vii) competes with the antibody CR3022; (viii) neutralizes one or more of SARS-CoV, SARS-CoV-2, MERS-CoV, HCoV-229E, HCoV-HKUl, HCoV-NL63, or
- compositions comprising at least one affinity-matured first anti-CoV-S antibody or antigen-binding antibody fragment and a pharmaceutically acceptable carrier or excipient.
- the at least one first antibody or antigen-binding antibody fragment may comprise: a VH comprising a VH CDR1, a VH CDR2, a VH CDR3; and a VL, comprising a VL CDR1 a VL CDR2, a VL CDR3, and the amino acid sequences of said VH CDR1, said VH CDR2, said VH CDR3, said VL CDR1, said VL CDR2, and said VL CDR3 are identical to the amino acid sequences of the VH CDR1, VH CDR2, VH CDR3, VL CDR1, VL CDR2, and VL CDR3, respectively, of an anti-CoV-S antibody selected from the group consisting of ADI-58120, ADI- 58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI- 58129, ADI-58130,
- the first antibody or antigen-binding antibody fragment may comprise an Fc region, optionally wherein the Fc region may comprise an amino acid sequence of SEQ ID NOs: 411, 412, 413, 414, 415, 416, or 417.
- the HC and LC of the first antibody or antigen-binding antibody fragment are the HC and LC, respectively, of an anti-CoV-S antibody selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, and ADI-58131.
- the composition may further comprise at least one second antibody or antigen-binding antibody fragment comprising a VH comprising a VH CDR1, a VH CDR2, a VH CDR3 and a VL, comprising a VL CDR1 a VL CDR2, a VL CDR3.
- the amino acid sequences of said VH CDR1, said VH CDR2, said VH CDR3, said VL CDR1, said VL CDR2, and said VL CDR3 may be identical to the amino acid sequences of the VH CDR1, VH CDR2, VH CDR3, VL CDR1, VL CDR2, and VL CDR3, respectively, of an anti-CoV-S antibody selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, and ADI-58131.
- the second antibody or antigen-binding antibody fragment may comprise an Lc region, optionally wherein the Lc region may comprise an amino acid sequence of SEQ ID NOs: 411, 412, 413, 414, 415, 416, or 417.
- the antibody or antigen-binding antibody fragment according to the present disclosure may comprise:
- a HC comprising the amino acid sequence of SEQ ID NO: 10 and a LC comprising the amino acid sequence of SEQ ID NO: 210;
- a HC comprising the amino acid sequence of SEQ ID NO: 50 and a LC comprising the amino acid sequence of SEQ ID NO: 250;
- a HC comprising the amino acid sequence of SEQ ID NO: 60 and a LC comprising the amino acid sequence of SEQ ID NO: 260;
- a HC comprising the amino acid sequence of SEQ ID NO: 70 and a LC comprising the amino acid sequence of SEQ ID NO: 270;
- a HC comprising the amino acid sequence of SEQ ID NO: 80 and a LC comprising the amino acid sequence of SEQ ID NO: 280; (9) a HC comprising the amino acid sequence of SEQ ID NO: 90 and a LC comprising the amino acid sequence of SEQ ID NO: 290;
- a HC comprising the amino acid sequence of SEQ ID NO: 110 and a LC comprising the amino acid sequence of SEQ ID NO: 310; or
- a HC comprising the amino acid sequence of SEQ ID NO: 120 and a LC comprising the amino acid sequence of SEQ ID NO: 320.
- composition according to the present disclosure may comprise: (A) at least one first antibody or antigen-binding antibody fragment selected from the group consisting of the antibodies or antigen-binding antibody fragments comprising the HC and LC combination as described above; and (B) a pharmaceutically acceptable carrier or excipient.
- the composition may additionally comprise at least one second antibody or antigen-binding antibody fragment selected from the group consisting of the antibodies or antigen-binding antibody fragments comprising the HC and LC combination as described above.
- the present disclosure further encompasses isolated antibodies and antigen binding antibody fragments thereof, which competes for binding with any one or more of the anti- CoV antibodies or antigen-binding antibody fragments thereof as described herein.
- the present disclosure also encompasses isolated antibodies or antigen-binding antibody fragments thereof, which bind the same epitope as any one or more of the anti-CoV antibodies or antigen-binding antibody fragments thereof as described herein.
- the present disclosure further encompasses affinity matured variants of any one or more of the anti-CoV antibodies or antigen-binding antibody fragments thereof as described herein.
- a method of treating a coronavirus infection by SARS-CoV, SARS-CoV-2, and/or another coronavirus optionally selected from the group consisting of MERS- CoV, HCoV-HKUl, HCoV-OC43, HCoV-229E, and HCoV-NL63 in a subject in need thereof comprising administering to the subject a therapeutically effective amount of an antibody, or antigen-binding antibody fragment thereof, which binds the same epitope as ADI-58125, and/or which competes for binding with ADI-58125.
- a method of decreasing the risk of mortality, hospitalization, mechanical ventilation, or a combination thereof in a patient infected by SARS-CoV, ARS-CoV-2, and/or another coronavirus optionally selected from the group consisting of MERS-CoV, HCoV- HKU1, HCoV-OC43, HCoV-229E, and HCoV-NL63 the method comprising administering to the subject a therapeutically effective amount of an isolated antibody, or antigen-binding antibody fragment thereof, which binds the same epitope as ADI-58125, and/or which competes for binding with ADI-58125.
- a method of preventing infection of a subject by SARS- CoV, SARS-CoV-2, and/or another coronavirus optionally selected from the group consisting of MERS-CoV, HCoV-HKUl, HCoV-OC43, HCoV-229E, and HCoV-NL63, the method comprising administering to the subject a therapeutically effective amount of an isolated antibody, or antigen binding antibody fragment thereof, which binds the same epitope as ADI-58125, and/or which competes for binding with ADI-58125.
- the present invention also provides a method for treating a coronavirus infection in a subject in need thereof, the method comprising administering to the subject an isolated antibody, or antigen-binding fragment thereof, wherein the antibody, or antigen-binding fragment thereof, comprises a heavy chain variable region (VH) comprising a VH CDR1 comprising SEQ ID NO:52, a VH CDR2 comprising SEQ ID NO:54, and a VH CDR3 comprising SEQ ID NO:56, and a light chain variable region (VL) comprising a VL CDR1 comprising SEQ ID NO:252, a VL CDR2 comprising SEQ ID NO:254, and a VL CDR3 comprising SEQ ID NO:256; and wherein the antibody, or antigen-binding fragment thereof, is administered at a dosage of at least 300 mg within 5, 4, 3, 2, or 1 days of symptom onset.
- VH heavy chain variable region
- VL light chain variable region
- the antibody, or antigen-binding fragment thereof is administered at a dosage of about 300 mg. In some embodiments, the antibody, or antigen-binding fragment thereof, is administered at a dosage of about 500 mg. In some embodiments, the antibody, or antigen-binding fragment thereof, is administered at a dosage of about 600 mg. In some embodiments, the antibody, or antigen-binding fragment thereof, is administered at a dosage of about 1200 mg. In some embodiments, the antibody, or antigen-binding fragment thereof, is administered at a dosage of about 4500 mg.
- the subject has at least one risk factor which renders them more prone to a poor clinical outcome.
- the at least one risk factor is selected from the group consisting of: an old age selected from the group consisting of over 55, over 60 or over 65 years old; diabetes, a chronic respiratory condition, obesity, hypertension, a cardiac or cardiovascular condition, a chronic inflammatory or autoimmune condition, and an immune compromised status.
- the subject is immunocompromised.
- the subject is at a high risk of disease progression.
- the subject is (a) age 50 years or above with no comorbid conditions or additional risk factors for progression of COVID-19; (b) between about 18 to about 50 years old and with one or more preexisting medical conditions selected from the group consisting of obesity, diabetes, chronic kidney disease, chronic lung disease, cardiac disease, sickle cell disease or thalassemia, solid organ or blood stem cell transplant recipient, other immunodeficiency due to underlying illness or immunosuppressant medication, Down Syndrome, stroke or cerebrovascular disease, substance use disorder, and pregnancy; or (c) between about 12 to about 17 years old and with one or more preexisting medical conditions selected from the group consisting of obesity, diabetes, chronic kidney disease, sickle cell disease or thalassemia, congenital or acquired heart disease, neurodevelopmental disorder, a medically-related technological dependence, asthma or chornic respiratory disease, solid organ or blood stem cell transplant recipient, other immunodeficiency due to underlying illness or immuno
- the subject is age 50 or above with no comorbid conditions or additional risk factors for progression of COVID-19. In some embodiments, the subject has hypertension with at least one medication prescribed or recommended. In some embodiments, the subject has moderate to severe asthma requiring daily therapy.
- the present invention provides a composition for use in inducing an immune response against CoV in a subject at high risk of COVID disease progression, wherein the composition comprises an antibody, or antigen-binding fragment thereof, comprises a heavy chain variable region (VH) comprising a VH CDR1 comprising SEQ ID NO:52, a VH CDR2 comprising SEQ ID NO:54, and a VH CDR3 comprising SEQ ID NO:56, and a light chain variable region (VL) comprising a VL CDR1 comprising SEQ ID NO:252, a VL CDR2 comprising SEQ ID NO:254, and a VL CDR3 comprising SEQ ID NO:256; and wherein the antibody, or antigen-binding fragment thereof, is administered within 5, 4 ,3, 2, or 1 days of symptom onset.
- VH heavy chain variable region
- VL light chain variable region
- the present invention provides a composition for use in treating or preventing a coronavirus infection in a subject at high risk of COVID disease progression, wherein the composition comprises an antibody, or antigen-binding fragment thereof, comprises a heavy chain variable region (VH) comprising a VH CDR1 comprising SEQ ID NO:52, a VH CDR2 comprising SEQ ID NO:54, and a VH CDR3 comprising SEQ ID NO:56, and a light chain variable region (VL) comprising a VL CDR1 comprising SEQ ID NO:252, a VL CDR2 comprising SEQ ID NO:254, and a VL CDR3 comprising SEQ ID NO:256; and wherein the antibody, or antigen-binding fragment thereof, is administered within 5, 4, 3, 2, or 1 days of symptom onset.
- VH heavy chain variable region
- VL light chain variable region
- the present invention provides a composition for use in treating a symptom of a coronavirus infection in a subject at high risk of COVID disease progression, wherein the composition comprises an antibody, or antigen-binding fragment thereof, comprises a heavy chain variable region (VH) comprising a VH CDR1 comprising SEQ ID NO:52, a VH CDR2 comprising SEQ ID NO:54, and a VH CDR3 comprising SEQ ID NO:56, and a light chain variable region (VL) comprising a VL CDR1 comprising SEQ ID NO:252, a VL CDR2 comprising SEQ ID NO:254, and a VL CDR3 comprising SEQ ID NO:256; and wherein the antibody, or antigen-binding fragment thereof, is administered within 5, 4, 3, 2, or 1 days of symptom onset.
- VH heavy chain variable region
- VL light chain variable region
- the present invention provides a composition for use in decreasing the risk of mortality, hospitalization, mechanical ventilation, or a combination thereof, in a subject at high risk of COVID disease progression, wherein the composition comprises an antibody, or antigen-binding fragment thereof, comprises a heavy chain variable region (VH) comprising a VH CDR1 comprising SEQ ID NO:52, a VH CDR2 comprising SEQ ID NO:54, and a VH CDR3 comprising SEQ ID NO:56, and a light chain variable region (VL) comprising a VL CDR1 comprising SEQ ID NO:252, a VL CDR2 comprising SEQ ID NO:254, and a VL CDR3 comprising SEQ ID NO:256; and wherein the antibody, or antigen-binding fragment thereof, is administered within 5, 4, 3, 2, or 1 days of symptom onset.
- VH heavy chain variable region
- VL light chain variable region
- Figure 1 depicts the mean serum concentration of ADI-58125 in 8 subjects following a single IM adminsitration of 300 mg ADI-58125 over a period of 21 days.
- Figure 2 depicts the quantitative systems pharmacology/physiologically based pharmacokinetic (QSP/PBPK) model-predicted and observed ADI-58125 concentrations following a 300 mg IM dose across the 21 -day sampling interval.
- the line represents the mediam model -predicted ADI-58125 profile, shaded area represents the 90% prediction interval, and black circles represent observed ADI-58125 concentrations.
- Figure 3 depicts the serum neutralizing titers from participants receiving a single 300 mg IM dose of ADI-58125.
- Figure 4 is a schematic depicting the experimental procedure for a plaque reduction neutralization test (PRNT)
- Figure 5 is a schematic depicting the experimental procedure for a post-infection assay to measure the concentration of antibody required to prevent viral replication in infected cells.
- Figure 6 depicts the concentration curves of ADI-58125 for preventing replication of SARS- CoV-2 D614G and beta B.1.351 variants in infected cells.
- Figure 7 depicts the neutralizing activity of ADI-58125 and ADI-58122 against authentic SARS-CoV-2 variants including the Alpha/B.1.1.7, the Beta/B.1.351, the Gamma/P.1, the Delta/B.1.617, and Omicron variants. Additional antibodies were also shown as comparators.
- Figure 8A depicts the neutralizing activity of ADI-58125 and ADI-58122 against pseudoviruses encoding spike proteins of circulating SARS-CoV-2 variants.
- Figure 8B depicts the neutralizing activity of ADI-58125 and ADI-58122 against pseudoviruses encoding SARS-CoV-2 spike proteins with sing!e-/doubIe-spike mutations, in a pseudovirus assay. The neutralizing activity was expressed as a fold change relative to the D614G reference variant.
- Figure 8C depicts the neutralizing activity of ADI-58125 against SARS-CoV-2 variants associated with resistance to EUA mAbs. The points represent neutralizing activity of ADI-58125 against SARS-CoV-2 variants.
- Figures 9A, 9B, 9C, and 9D depict an overview of QSP/PBPK model.
- Figure 9A depicts a tissue-level diagram.
- Figure 9B depicts a cellular-level diagram specifically for the upper airway, lower airway, and alveolar spaces within the lung.
- Figure 9C depicts the predicted median ADI- 58125 concentration in NHP serum following a single ADI-58125 10 mg/kg IM dose with observed NHP ADI-58125 concentration data overlaid (black dots). The shaded area represents the 90% prediction interval.
- Figure 9D depicts the predicted median human serum and ELF PK in humans following a single ADI-58125 300 mg IM dose with observed human serum (red line) ADI-58125 concentration data overlaid (black dots), ELF upper airway (blue line), and ELF lower airway (gold). The shaded area represents the 90% prediction interval.
- CL up rate of pinocytosis of antibody entry and exit from the endosomal space
- CL up-epi rate of pinocytosis of antibody entry and exit from the epithelial space
- FR fraction of FcRn bound antibody that recycles to the vascular space
- L lymphatic flow rate
- LG large, k de , degradation constant
- k 0ff first-order dissociation rate constant of antibody from FcRn
- k on second-order association rate constant for binding of antibody to FcRn
- Q blood or tissue flow rate
- SM small.
- Figures 10A-10B depict a QSP/PBPK model forecast of ADI-58125 300 mg IM in adults.
- Figure 10A depicts the QSP/PBPK predicted median ADI-58125 concentration following a single 300 mg IM injection in different compartments overlaid by the threashold associated with 95% viral growth suppression (as indicated by the dotted line) for the SARS-CoV-2 Beta variant in a post infection assay. The shaded area represents the 90% prediction interval.
- Figure 10B depicts the receptor occupancy (RO) of ADI-58125 at various Delta variant virion densities (10 7 , 10 s and 10 9 ) after a single 300 mg IM injection expressed as percent occupancy with the dotted line representing the threshold for 90% RO.
- RO receptor occupancy
- Figure 11 is a schematic representation for the in vivo efficacy study in hamsters.
- Hamsters were treated with a range of ADI-58125 doses (9.25 - 2000 ⁇ g) or control mAh 24 hours prior to challenge with SARS-2/WA-1 to evaluate prophylactic efficacy of ADI-58125.
- Figures 12A-12B depict the impact of ADI-58125 on weight gain and lung pathology in SARS-CoV-2 -infected Syrian hamsters. Specifically, Figure 12A depicts the changes in weight from baseline on Day 6. Figure 12B depicts the total lung pathology score. Bars represent mean ⁇ standard deviation. Dotted line represents no change in weight from baseline. Statistical comparison was calculated using unpaired, two-sided t-tests (A) and unpaired two-sided Mann-Whiteney tests (B): *P ⁇ 0.05; **** P ⁇ 0.0001 vs controls.
- Figure 13 depicts the impact of ADI-58125 on viral load in SARS-CoV-2 -infected Syrian hamsters. Bars represent mean ⁇ standard deviation. Dotted line represents limit of detection. Statistical comparison was calculated using unpaired, two-sided t-tests: **P ⁇ 0.005; *** P ⁇ 0.001; ****p ⁇ 0.0001 vs controls.
- Figure 14 is a schematic representation for the in vivo efficacy study in non-human primates (NHP). Rhesus macaques were treated with ADI-58125 at 5 mg/kg or 25 mg/kg, or control mAh (25 mg/kg) 3 days prior to challenge with SARS-2/WA-1 to evaluate prophylactic efficacy of ADI-58125.
- Figure 15 depicts the impact of ADI-58125 on viral replication in Rhesus macaques, specifically, the impact on replication of sub-genomic viral RNA in bronchoalveolar lavage (BAL), nasopharyngeal (NP) and oropharyngeal (OP) samples.
- BAL bronchoalveolar lavage
- NP nasopharyngeal
- OP oropharyngeal
- Figure 16 depicts the impact of ADI-58125 on viral titre in Rhesus macaques.
- Figure 17 depicts the mean serum concentration of ADI-58125 in 8 subjects following a single IM adminsitration of 300 mg ADI-58125 over a period of 3 months.
- Figure 18 depicts the serum neutralizing titers from participants receiving a single 300 mg IM dose of ADI-58125 as compared to peak responses following AZD1222 and mRNA-1273 vaccination.
- Statistical analysis is by 2-tailed Mann-Whitney U test (*P ⁇ 0.05; **P ⁇ 0.01; ***P ⁇ 0.0001).
- Figure 19 depicts the mean serum concentration of ADI-58125 in subjects following a single IM adminsitration of 300 mg ADI-58125, a single IM adminsitration of 600 mg ADI-58125, and a single IV administration of 500 mg ADI-58125 over a period of 6 months.
- Figure 20 depicts the relationship between ADI-58125 serum concentration and sVNA over a period of 6 months.
- the line represents the mean linear regression. The hading represents 95% confidence interval.
- Black circles represent individual time matched ADI-58125 serum concentration and MN50 sVNA titer measurements. Linear regression excluded samples that were taken following SARS-CoV-2 vaccination from participants who received vaccination during the trial.
- Figures 21A, 21B, and 21C depict the simulated human body weight (A) and distribution of estimated K D,FcRn values for other extended half-life mAbs in healthy human adults (log-normal, B) and NHPs (normal, C).
- Figures 22A-22B depict the observed (dots) and model-forecasted NHP median (90% prediction interval) serum ADI-58125 PK profiles based on distribution of NHP K D,FcRn values for other extended half-life mAbs following intravenous (IV; A) and intramuscular (IM; B) administration. Dots represent the raw observed data. Solid lines represent simulated medians. The shaded area represents the 90% prediction interval.
- Figures 23A-23B depict the observed (dots) and optimized QSP model-predicted NHP median (90% prediction interval) serum ADI-58125 concentration-time profiles in NHPs following intravenous (IV; A) and intramuscular (IM; B) administration. Dots represent the raw observed data. Solid lines represent simulated medians. The shaded area represents the 90% prediction interval.
- Figures 24A-24D depict the observed (dots) versus QSP model-predicted median (90% prediction interval) serum ADI-58125 PK profiles in healthy adult participants predicted a priori based on the distribution of human K D,FcRn values for other extended half-life mAbs (A, C) and after optimization (B, D). Dots represent the raw observed data. Solid lines represent simulated medians. The shaded area represents the 90% prediction interval.
- Figures 25A-25C depict the QSP model-predicted median (90% prediction interval) serum ADI-58125 PK profiles following a single IM 150 mg (A), 300 mg (B), and 450 mg (C) injection in humans predicted a priori based on the distribution of human K D,FcRn values for other extended half- life mAbs (as shown in Figures 21 A-B). Dashed line represents 100 * in vitro IC 90 of 0.011 ⁇ g/mL or 1.1 mg/L against the USA-WA1/2020 variant.
- Figures 26A-26C depict the optimized QSP model-predicted median (90% prediction interval) serum ADI-58125 PK profiles following a single IM 150 mg (A), 300 mg (B), and 450 mg (C) injection in humans. Dashed line represents 100 * in vitro IC 90 of 0.011 ⁇ g/mL or 1.1 mg/L against the USA-WA1/2020 variant.
- Figure 27 depicts the serum neutralizing titers (geometric mean MN80) from participants receiving a single 300 mg IM dose of ADI-58125 as compared to peak titers with 2-dose mRNA-1273 vaccination. “Statistical comparisons for Delta were based on prelilminary results. b Peak vaccine response timepoint: 7 to 31 days post 2nd dose; c Exeludes samples taken following SARS-CoV-2 vaccination. Statistical comparisons of titres achieved by ADI-58125 against each variant were performed using 2-sided Mann Whitney U tests: *P ⁇ 0.05; **P ⁇ 0.01; ***P ⁇ 0.001; ns, non-significant vs mRNA-1273 vaccination response against the corresponding variant.
- Figure 28 depicts the visual predictive check plots for the base ADI-58125 pharmacokinetic model.
- Figure 29A depicts the key binding residues in SARS-CoV-2 S protein for ADI-58125, and the strong conservation of these residues among global circulating SARS-CoV-2 variants.
- FIG. 29B depicts the key binding residues in SARS-CoV-2 S protein for class 1 (e.g., LY-C0VOI6, REGN10933), class 2 (e.g., P2B-2F6, LY- CoV555, COV2-2196) and class 3 (e.g., REGN10987, COV2-2130, LY-CoV1404) antibodies.
- class 1 e.g., LY-C0VOI6, REGN10933
- class 2 e.g., P2B-2F6, LY- CoV555, COV2-2196
- class 3 e.g., REGN10987, COV2-2130, LY-CoV1404
- Figure 30 depicts that the newly emerged Omicron (B.1.1.529) variant contains mutations in the epitopes of all three public classes of antibodies.
- Figure 31 depicts that ADI-58125 binds to an epitope distinct from those recognized by common elicited nAbs.
- Figure 32A depicts the non-overlapping epitopes between ADI-58125 and other clinical stage/EUA authorized antibodies.
- Figure 32B depicts the non-overlapping epitopes between ADI- 58122 and ADI-58125.
- Figure 33 depicts the locally extensive area of red discoloration and consolidation (black arrows) in the hilar region of the right caudal lung lobe at 6 dpi. RM 1221.
- Figure 34 depicts the cut section of right caudal lung lobe showing the locally extensive region of consolidation/non-inflating (black arrow) at 6 dpi. RM 1221.
- Figures 35A-35E depict the right caudal lung lobe, central section.
- Figure 35A Multifocal moderate interstitial pneumonia at 6 dpi, 2x, H&E, RM 1221.
- Figure 35B boxed area from Figure 35A, Moderate inflammation expanding the alveolar septa and extending into the alveolar lumina and moderate type II pneumocyte hyperplasia at 6 dpi, lOx, H&E, RM 1221.
- Figure 35C boxed area from Figure 35B, alveolar septa were expanded by moderate numbers of lymphocytes, histiocytes, neutrophils and fibrin, which extended into lumen.
- Multifocally alveolar septa were lined by type II pneumocytes (arrows) at 6 dpi, 20x, H&E, RM 1221 Figure 35D. Rare IHC positive labeling in areas of inflammation, 2x, nucleocapsid protein for SARS-CoV-2, RM 1221. Figure 35E, boxed areas from Figure 35D, rare IHC positive labeling in areas of inflammation, 20x, nucleocapsid protein for SARS- CoV-2, RM 1221.
- Figures 36A-36B depict nasal turbinate.
- Figure 36A Locally extensive region of ulceration with subadjacent inflammation, hemorrhage and edema at 5 dpi, 4x, H&E, RM 7137.
- Figure 36B boxed area from Figure 36A: Mucosal ulceration with mild to moderate lymphoplasmacytic and neutrophilic inflammation with necrosis, hemorrhage and edema at 5 dpi, lOx, H&E, RM 7137.
- Figures 37A-37D depict right caudal lung lobe, central section.
- Figure 37A Multifocal mild inflammation surrounding blood vessels, expanding alveolar septa and occasionally extending into the lumina at 5 dpi, 2x, H&E, RM 7175.
- Figure 37B boxed area from Figure 37A: Mild inflammation composed predominately of histiocytes and lymphocytes with fewer neutrophils, around vessels and expanding septa at 5 dpi, lOx, H&E, RM 7175.
- Figure 37C IHC labeling was not identified in areas of inflammation at 5 dpi, 2x, nucleocapsid protein for SARS-CoV-2, RM 7175.
- Figure 37D IHC labeling was not identified in areas of inflammation at 5 dpi, lOx, nucleocapsid protein for SARS- CoV-2, RM 7175.
- Figures 38A-38D depict lung, left caudal lobe, central section. RM 1430.
- Figure 38A Multifocal mild inflammation surrounding blood vessels, expanding alveolar septa and extending into the alveolar lumina at 6 dpi, 2x, H&E, RM 1430.
- Figure 38B boxed area from Figure 38A: Mild inflammation composed predominately of histiocytes and lymphocytes with fewer neutrophils, around vessels and expanding septa and within the alveolar lumina at 6 dpi, lOx, H&E, RM 1430.
- Figure 38C Figure 38C.
- IHC labeling was not identified in areas of inflammation at 6 dpi, 2x, nucleocapsid protein for SARS-CoV-2, RM 1430.
- Figure 38D IHC labeling was not identified in areas of inflammation at 6 dpi, lOx, nucleocapsid protein for SARS-CoV-2, RM 1430.
- Figure 39 depicts mild red discoloration for all lung lobes at 5 dpi. RM 1294.
- Figure 40 depicts multifocal mild red discoloration throughout all lobes with locally extensive fibrinous adhesions noted from the left caudal lung lobe to the thoracic cavity at 5 dpi. RM 5240.
- Figure 41 depicts average fold change in IC50 for ADI-58125 relative to the D614G reference strain in a lentiviral pseudovirus assay.
- Figure 42A shows the neutralization curves for ADI-58125 and ADI-58122 against Omicron BA.l variant (red) and the D614G reference strain (black) using a pseudo virus neutralization assay.
- Figue 42B shows the neutralization curves for ADI-58125 against D614G and Omicron sublineages (BA.l, BA.1.1 and BA.2) using a pseudovirus neutralization assay.
- Figure 43 shows the neutralization curves for ADI-58125, ADI-58122, ADI-58127 and other commercial antibodies against Victoria and Omicron sublineages (BA.l, BA.1.1 and BA.2) in an authentic neutralization assay.
- Figure 44 shows the neutralization data for ADI-58125, ADI-58122, ADI-58127 and other commercial antibodies against Victoria and all variants of concern, Alpha, Beta, Gamma, Delta and Omicron (BA.l., BA.1.1, BA.2) in an authentic neutralization assay.
- Figure 45 depicts the correlation between serum neutralizing antibody titers induced by vaccination or mAh administration and reported vaccine or mAh efficacy against symptomatic COVID-19. Geometric mean serum neutralizing titers for samples obtained from AZD1222 and BNT162b2 vaccinees at the indicated timepoints against the indicated variants are plotted against reported vaccine efficacy at matched timepoints against the indicated variants. Red and black dotted lines indicate predicted month 3 serum neutralizing antibody titers against the Omicron variant following administration of 300 mg IM AZD7442 or 1200 mg IM ADI-58125, respectively.
- Figure 46 depicts the predicted median (10%-90% PI) ADI-58125 serum concentration-time profile following single 1200 mg IM dose. Simulation from preliminary PopPK model developed using data from the first-in-human study. Solid black line and shaded area represent median and 80% (10%-90%) prediction interval, respectively. Solid red line indicates the median serum concentration of ADI-58125 associated with the target threshold serum neutralizing titer of 1:100. Blue and green dashed/dotted lines represent the median IC90 (dashed) and IC50 (dotted) values for ADI-58125 against the Omicron variant in the ELF assuming an ELF: serum ratio of 10% (blue) or 15% (green).
- ELF epidermal lining fluid
- sVNT serum virus neutralizing titer
- Figure 47 depicts the predicted median (10%-90% PI) ADI-58125 serum concentration-time profile following single 1200 mg IV dose. Simulation from preliminary PopPK model developed using data from the first-in-human study. Solid black line and shaded area represent median and 80% (10%-90%) prediction interval, respectively. Solid red line indicates the median serum concentration of ADI-58125 associated with the target threshold serum neutralizing titer of 1:100.
- Figure 48 depicts the neutralization curves of authentic Omicron virus by ADI-58125 and EUA mAbs. Antibody neutralization of authentic Omicron virus (B.1.1.529), as determined by a focus reduction neutralization assay. Data points and error bars represent the mean and standard deviation. Dotted lines indicate 50% and 90% neutralization.
- Figure 49 depicts the goodness-of-fit plots for the ADI-58125 PK model.
- Figure 50 depicts the key ADI-58125 PK parameter distributions across all doses (300mg IM, 500 mg IV, and 600 mg IM).
- CL clearance.
- Vss steady state volume of distribution.
- Figure 51 depicts the population median predicted concentrations of ADI-58125 over time with IV and IM administration.
- Figure 52A provides a graph comparing binding affinity of ADI-58125 and affinity maturation progenies for the SI from the Omicron BA.l varaint.
- Figure 52B provides a graph comparing the ability of ADI-58125 and affinity maturation progenies to neutralize the Omicron BA.l varaint.
- Figure 53A provides a graph comparing binding affinity of affinity maturation ADI-58125 progenies for different variants of concern.
- Figure 53B provides a graph comparing the ability of affinity maturation ADI-58125 progenies to neutralize different variants of concern.
- Figure 54 depicts the median (90% confidenence interval) ADI-58125 concentration-time profile following a 300 mg IM dose for phase 1 and phase 2/3 prevention and treatment studies. Shaded areas represent 90% CL Solid line represents median.
- Figure 55 depicts the correlation between serum neutralizing antibody titers induced by vaccination or mAh administration and reported vaccine or mAh efficacy against symptomatic COVID-19. Geometric mean serum neutralizing titers for samples obtained from AZD1222 and BNT162b2 vaccinees at the indicated timepoints against the indicated variants are plotted against reported vaccine efficacy at matched timepoints against the indicated variants.
- Serum concentrations from population PK model based on Phase 2/3 data of ADI-58125 on days 55 and 76 (Omicron BA.1/BA1.1), and 90 (Delta) were used to calculate predictive nAb titers and corresponding efficacy for the Delta and Omicron variants and then compared to clinical efficacy results from the EVADE clinical trial in prevention of COVID-19.
- Abbreviations: FRNT50 50% focus reduction neutralization test.
- Spike protein S protein includes any coronavirus form of S protein.
- coronavirus S protein (“CoV-S”) is used to describe the S protein of any coronaviruses.
- the “SARS-CoV-S” and “SARS-CoV-2-S” encompass the S protein of SARS-CoV and of SARS-CoV-2.
- SEQ ID NO: 401 is an exemplary polypeptide sequence of SARS-CoV-S, comprising 1288 amino acids (Accession# PDB: 6VSB_B).
- SEQ ID NO: 403 is an exemplary polypeptide sequence of SARS-CoV-2-S, comprising 1273 amino acids (GenBank: QHD43416.1).
- SEQ ID NO: 402 (3864 nucleotides) encodes the SARS-CoV-S (SEQ ID NO: 401) and SEQ ID NO: 404 (3822 nucleotides, NC_045512:21563.25384, also see the corresponding region of GenBank: MN908947) encodes SARS-CoV -2-S (SEQ ID NO: 403).
- the “SARS-CoV-S” and “SARS-CoV -2-S” encompass any mutants, splice variants, isoforms, orthologs, homologs, and variants of SEQ ID Nos 401 and 403.
- the CoV-S comprises a polypeptide sequence having at least 85%, 90%, 95%, 96%, 97%, 98%, or 99% identity to either SEQ ID NO: 401 or SEQ ID NO: 403.
- Effective treatment or prevention of CoV infection refers to eliminating CoV from the subject or preventing the expansion of CoV in the subject or eliminating or reducing the symptoms such as fever, cough, shortness of breath, runny nose, congestion, conjunctivitis, and/or gastrointestinal symptoms after administration of an effective amount of an anti-CoV-S antibody or antigen-binding fragment thereof.
- effective treatment may eliminate the need for the subject to be placed on a ventilator or reduce the time the subject needs to be on a ventilator.
- the treatment may be effected as a monotherapy or in association with another active agent such as an antiviral agent or anti-inflammatory agent by way of example.
- beneficial or desired clinical results include, but are not limited to, one or more of the following: improvement in any aspect of COV-S-related conditions such as fever or cough.
- beneficial or desired clinical results include, but are not limited to, one or more of the following: improvement in any aspect of COV-S-related conditions such as fever or cough.
- this includes lessening severity, alleviation of fever, cough, shortness of breath, and other associated symptoms, reducing frequency of recurrence, increasing the quality of life of those suffering from the CoV-related symptoms, and decreasing dose of other medications required to treat the CoV-related symptoms.
- Other associated symptoms include, but are not limited to, diarrhea, conjunctivitis, loss of smell, and loss of taste.
- Still other symptoms which may be alleviated or prevented include inflammation, cytokine storm and/or sepsis.
- Reducing incidence or “prophylaxis” or “prevention” means any of reducing severity for a particular disease, condition, symptom, or disorder (the terms disease, condition, and disorder are used interchangeably throughout the application).
- Reduction in severity includes reducing drugs and/or therapies generally used for the condition by, for example, reducing the need for, amount of, and/or exposure to drugs or therapies.
- Reduction in severity also includes reducing the duration, and/or frequency of the particular condition, symptom, or disorder (including, for example, delaying or increasing time to next episodic attack in an individual). This further includes eliminating the need for the subject to be placed on a ventilator or reducing the time the subject needs to be on a ventilator.
- “Ameliorating" one or more symptoms of CoV infection-related conditions means a lessening or improvement of one or more symptoms of the condition, e.g., fever or cough or shortness of breath as compared to not administering an anti-CoV-S antagonist antibody. "Ameliorating” also includes shortening or reduction in duration of a symptom. Again, this may include eliminating the need for the subject to be placed on a ventilator or reducing the time the subject needs to be on a ventilator.
- “controlling CoV-related symptom” or “controlling” another CoV-S-related condition refers to maintaining or reducing severity or duration of one or more symptoms of the condition (as compared to the level before treatment).
- the duration or severity or frequency of symptoms is reduced by at least about any of 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100% in the individual as compared to the level before treatment.
- the reduction in the duration or severity, or frequency of symptoms can last for any length of time, e.g., 2 weeks, 4 weeks (1 month), 8 weeks (2 months), 16 weeks (3 months), 4 months, 5 months, 6 months, 9 months, 12 months, etc.
- delaying means to defer, hinder, slow, retard, stabilize, and/or postpone progression of the condition or disease.
- This delay can be of varying lengths of time, depending on the history of the condition or disease and/or individuals being treated.
- a sufficient or significant delay can, in effect, encompass prevention, in that the individual does not develop symptoms.
- a method that "delays" development of the symptom is a method that reduces probability of developing the symptom in a given time frame and/or reduces extent of the symptoms in a given time frame, when compared to not using the method. Such comparisons are typically based on clinical studies, using a statistically significant number of subjects.
- "Development” or “progression” of a CoV-related condition such as cough or fever means initial manifestations and/or ensuing progression of the disorder. Development of cough or fever can be detectable and assessed using standard clinical techniques as well known in the art. However, development also refers to progression that may be undetectable. For purpose of this disclosure, development, or progression refers to the biological course of the symptoms.
- “Development” includes occurrence, recurrence, and onset. As used herein "onset” or "occurrence” of a condition includes initial onset and/or recurrence.
- an "effective dosage” or “effective amount” of drug, compound, or pharmaceutical composition is an amount sufficient to effect beneficial or desired results.
- beneficial or desired results include results such as eliminating or reducing the risk, lessening the severity, or delaying the outset of the disease, including biochemical, histological, and/or behavioral symptoms of the disease, its complications and intermediate pathological phenotypes presenting during development of the disease.
- beneficial or desired results include clinical results such as reducing symptom intensity, duration, or frequency, and decreasing one or more symptoms resulting from CoV infection, including its complications and intermediate pathological phenotypes presenting during development of the disease, increasing the quality of life of those suffering from the disease, decreasing the dose of other medications required to treat the disease, enhancing effect of another medication, and/or delaying the progression of the disease of patients, eliminating the need for the subject to be placed on a ventilator or reducing the time the subject needs to be on a ventilator.
- an effective dosage can be administered in one or more administrations.
- an effective dosage of drug, compound, or pharmaceutical composition is an amount sufficient to accomplish prophylactic or therapeutic treatment either directly or indirectly.
- an effective dosage of a drug, compound, or pharmaceutical composition may or may not be achieved in conjunction with another drug, compound, or pharmaceutical composition.
- an "effective dosage" may be considered in the context of administering one or more therapeutic agents, and a single agent may be considered to be given in an effective amount if, in conjunction with one or more other agents, a desirable result may be or is achieved.
- a “suitable host cell” or “host cell” generally includes any cell wherein the subject anti-CoV- S antibodies and antigen-binding fragments thereof can be produced recombinantly using techniques and materials readily available.
- the anti-CoV-S antibodies and antigen-binding fragments thereof of the present disclosure can be produced in genetically engineered host cells according to conventional techniques.
- Suitable host cells are those cell types that can be transformed or transfected with exogenous DNA and grown in culture, and include bacteria, fungal cells (e.g., yeast), and cultured higher eukaryotic cells (including cultured cells of multicellular organisms), particularly cultured mammalian cells, e.g., human or non-human mammalian cells.
- these antibodies may be expressed in CHO cells.
- Techniques for manipulating cloned DNA molecules and introducing exogenous DNA into a variety of host cells are disclosed by Sambrook et ai, Molecular Cloning: A Laboratory Manual, 2nd ed., Cold Spring Harbor, N.Y.: Cold Spring Harbor Laboratory Press (1989), and Current Protocols in Molecular Biology, Ausubel et ai, editors, New York, NY: Green and Wiley and Sons (1993).
- the antibodies may be expressed in mating competent yeast, e.g., any haploid, diploid or tetraploid yeast that can be grown in culture.
- Yeast useful in fermentation expression methods may exist in a haploid, diploid, or other polyploid form.
- a “selectable marker” herein refers to a gene or gene fragment that confers a growth phenotype (physical growth characteristic) on a cell receiving that gene as, for example through a transformation event.
- the selectable marker allows that cell to survive and grow in a selective growth medium under conditions in which cells that do not receive that selectable marker gene cannot grow.
- Selectable marker genes generally fall into several types, including positive selectable marker genes such as a gene that confers on a cell resistance to an antibiotic or other drug, temperature when two temperature sensitive (“ts”) mutants are crossed or a ts mutant is transformed; negative selectable marker genes such as a biosynthetic gene that confers on a cell the ability to grow in a medium without a specific nutrient needed by all cells that do not have that biosynthetic gene, or a mutagenized biosynthetic gene that confers on a cell inability to grow by cells that do not have the wild type gene; and the like.
- positive selectable marker genes such as a gene that confers on a cell resistance to an antibiotic or other drug, temperature when two temperature sensitive (“ts”) mutants are crossed or a ts mutant is transformed
- negative selectable marker genes such as a biosynthetic gene that confers on a cell the ability to grow in a medium without a specific nutrient needed by all cells that do not have that biosynthetic gene,
- An “expression vector” herein refers to DNA vectors containing elements that facilitate manipulation for the expression of a foreign protein within the target host cell, e.g., a bacterial, insect, yeast, plant, amphibian, reptile, avian, or mammalian cell, e.g., a CHO or HEK cell.
- a foreign protein within the target host cell
- a bacterial host e.g. E. coli
- vectors will include sequences to facilitate such manipulations, including a bacterial origin of replication and appropriate bacterial selection marker. Selection markers encode proteins necessary for the survival or growth of transformed host cells grown in a selective culture medium.
- Selection genes encode proteins that (a) confer resistance to antibiotics or other toxins, (b) complement auxotrophic deficiencies, or (c) supply critical nutrients not available from complex media.
- Exemplary vectors and methods for transformation of yeast are described, for example, in Burke, D., Dawson, D., & Stearns, T., Methods in yeast genetics: a Cold Spring Harbor Laboratory course manual, Plainview, NY : Cold Spring Harbor Laboratory Press (2000).
- Expression vectors for use in the methods of the disclosure may include yeast or mammalian specific sequences, including a selectable auxotrophic or drug marker for identifying transformed host s.
- a drug marker may further be used to amplify copy number of the vector in a yeast host cell.
- the polypeptide coding sequence of interest is operably linked to transcriptional and translational regulatory sequences that provide for expression of the polypeptide in the desired host cells, e.g., yeast or mammalian cells.
- These vector components may include, but are not limited to, one or more of the following: an enhancer element, a promoter, and a transcription termination sequence. Sequences for the secretion of the polypeptide may also be included, e.g. a signal sequence, and the like.
- An origin of replication e.g., a yeast or mammalian origin of replication, is optional, as expression vectors may be integrated into the host cell genome.
- Nucleic acids are "operably linked" when placed into a functional relationship with another nucleic acid sequence.
- DNA for a signal sequence is operably linked to DNA for a polypeptide if it is expressed as a preprotein that participates in the secretion of the polypeptide; a promoter or enhancer is operably linked to a coding sequence if it affects the transcription of the sequence.
- "operably linked” means that the DNA sequences being linked are contiguous, and, in the case of a secretory leader, contiguous and in reading frame. However, enhancers do not have to be contiguous.
- Linking is accomplished by ligation at convenient restriction sites or via a PCR/recombination method familiar to those skilled in the art (GATEWAY ® Technology (universal method for cloning DNA); Invitrogen, Carlsbad California). If such sites do not exist, the synthetic oligonucleotide adapters or linkers are used in accordance with conventional practice.
- Promoters are untranslated sequences located upstream (5') to the start codon of a structural gene (generally within about 100 to 1000 bp) that control the transcription and translation of particular nucleic acid sequences to which they are operably linked. Such promoters fall into several classes: inducible, constitutive, and repressible promoters (that increase levels of transcription in response to absence of a repressor). Inducible promoters may initiate increased levels of transcription from DNA under their control in response to some change in culture conditions, e.g., the presence or absence of a nutrient or a change in temperature.
- the promoter fragment may also serve as the site for homologous recombination and integration of the expression vector into the same site in the host cell, e.g., yeast or mammalian cell, genome; alternatively, a selectable marker may be used as the site for homologous recombination.
- a selectable marker may be used as the site for homologous recombination.
- Suitable promoters for use in different eukaryotic and prokaryotic cells are well known and commercially available.
- the polypeptides of interest may be produced recombinantly not only directly, but also as a fusion polypeptide with a heterologous polypeptide, e.g. a signal sequence or other polypeptide having a specific cleavage site at the N-terminus of the mature protein or polypeptide.
- a heterologous polypeptide e.g. a signal sequence or other polypeptide having a specific cleavage site at the N-terminus of the mature protein or polypeptide.
- the signal sequence may be a component of the vector, or it may be a part of the polypeptide coding sequence that is inserted into the vector.
- the heterologous signal sequence selected preferably is one that is recognized and processed through one of the standard pathways available within the host cell, e.g., a mammalian cell, an insect cell, or a yeast cell.
- secretion signals of interest also include mammalian and yeast signal sequences, which may be heterologous to the protein being secreted, or may be a native sequence for the protein being secreted.
- Signal sequences include pre peptide sequences, and in some instances may include propeptide sequences.
- Many such signal sequences are known in the art, including the signal sequences found on immunoglobulin chains, e.g., K28 preprotoxin sequence, PHA-E, FACE, human MCP-1, human serum albumin signal sequences, human Ig heavy chain, human Ig light chain, and the like. For example, see Hashimoto et. al., Protein Eng., 11(2):75 (1998); and Kobayashi et. al., Therapeutic Apheresis, 2(4):257 (1998)).
- Transcription may be increased by inserting a transcriptional activator sequence into the vector.
- These activators are cis-acting elements of DNA, usually about from 10 to 300 bp, which act on a promoter to increase its transcription.
- Transcriptional enhancers are relatively orientation and position independent, having been found 5' and 3' to the transcription unit, within an intron, as well as within the coding sequence itself. The enhancer may be spliced into the expression vector at a position 5' or 3' to the coding sequence, but is preferably located at a site 5' from the promoter.
- Expression vectors used in eukaryotic host cells may also contain sequences necessary for the termination of transcription and for stabilizing the rnRNA. Such sequences are commonly available from 3' to the translation termination codon, in untranslated regions of eukaryotic or viral DNAs or cDNAs. These regions contain nucleotide segments transcribed as polyadenylated fragments in the untranslated portion of the rnRNA.
- Plasmids from the transformants are prepared, analyzed by restriction endonuclease digestion, and/or sequenced.
- recombination methods based on specific attachment (“ att ”) sites and recombination enzymes may be used to insert DNA sequences into a vector.
- att specific attachment
- Such methods are described, for example, by Landy, Ann. Rev. Biochem., 58:913-949 (1989); and are known to those of skill in the art.
- Such methods utilize intermolecular DNA recombination that is mediated by a mixture of lambda and E. coli -encoded recombination proteins. Recombination occurs between att sites on the interacting DNA molecules.
- Att sites may be introduced into a sequence of interest by ligating the sequence of interest into an appropriate vector; generating a PCR product containing att B sites through the use of specific primers; generating a cDNA library cloned into an appropriate vector containing att sites; and the like.
- Folding refers to the three-dimensional structure of polypeptides and proteins, where interactions between amino acid residues act to stabilize the structure. While non-covalent interactions are important in determining structure, usually the proteins of interest will have intra- and/or intermolecular covalent disulfide bonds formed by two cysteine residues. For naturally occurring proteins and polypeptides or derivatives and variants thereof, the proper folding is typically the arrangement that results in optimal biological activity, and can conveniently be monitored by assays for activity, e.g. ligand binding, enzymatic activity, etc.
- the expression host may be further modified by the introduction of sequences encoding one or more enzymes that enhance folding and disulfide bond formation, i.e. foldases, chaperonins, etc.
- sequences may be constitutively or inducibly expressed in the host cell, using vectors, markers, etc. as known in the art.
- sequences, including transcriptional regulatory elements sufficient for the desired pattern of expression are stably integrated in the yeast genome through a targeted methodology.
- the eukaryotic protein disulfide isomerase (“PDI”) is not only an efficient catalyst of protein cysteine oxidation and disulfide bond isomerization, but also exhibits chaperone activity. Co-expression of PDI can facilitate the production of active proteins having multiple disulfide bonds. Also of interest is the expression of immunoglobulin heavy chain binding protein (“BIP”); cyclophilin; and the like.
- BIP immunoglobulin heavy chain binding protein
- Cultured mammalian cells are exemplary hosts for production of the disclosed anti-CoV-S antibodies and antigen-binding fragments thereof.
- CHO cells are particularly suitable for expression of antibodies.
- Many procedures are known in the art for manufacturing monoclonal antibodies in mammalian cells. (See, Galfre, G. and Milstein, C., Methods Enzym., 73:3-46, 1981; Basalp et al, Turk. J. Biol., 24:189-196, 2000; Wurm, F.M., Nat. Biotechnol., 22:1393-1398, 2004; and Li et al, mAbs, 2(5):466-477, 2010).
- common host cell lines employed in mammalian monoclonal antibody manufacturing schemes include, but are not limited to, human embryonic retinoblast cell line PER.C6® (Crucell N.V., Leiden, The Netherlands), NSO murine myeloma cells (Medical Research Council, London, UK), CV 1 monkey kidney cell line, 293 human embryonic kidney cell line, BHK baby hamster kidney cell line, VERO African green monkey kidney cell line, human cervical carcinoma cell line HELA, MDCK canine kidney cells, BRL buffalo rat liver cells, W138 human lung cells, HepG2 human liver cells, MMT mouse mammary tumor cells, TRI cells, MRC5 cells, Fs4 cells, myeloma or lymphoma cells, or Chinese Hamster ( Cricetulus griseus ) Ovary (CHO) cells, and the like.
- human embryonic retinoblast cell line PER.C6® Crucell N.V., Leiden, The Netherlands
- NSO cells are a non-Ig secreting, non-light chain- synthesizing subclone of NS-1 cells that are resistant to azaguanine.
- CHO-DXB11 CHO-DUKX
- CHO- pro3, CHO-DG44 CHO 1-15
- CHO DP-12 CHO DP-12
- Lec2, M1WT3, Lec8, ⁇ gsA-745 and the like, ah of which are genetically altered to optimize the cell line for various parameters.
- Monoclonal antibodies are commonly manufactured using a batch fed method whereby the monoclonal antibody chains are expressed in a mammalian cell line and secreted into the tissue culture medium in a bioreactor. Medium (or feed) is continuously supplied to the bioreactor to maximize recombinant protein expression. Recombinant monoclonal antibody is then purified from the collected media.
- nucleic acids encoding the antibody or fragment thereof are generally inserted into a replicable vector for further cloning (amplification of the DNA) or for expression.
- DNA encoding the antibody is readily isolated or synthesized using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to DNAs encoding the heavy and light chains of the antibody).
- the vector components generally include, but are not limited to, one or more of the following: a signal sequence, an origin of replication, one or more marker genes, an enhancer element, a promoter, and a transcription termination sequence. Selection of promoters, terminators, selectable markers, vectors, and other elements is a matter of routine design within the level of ordinary skill in the art. Many such elements are known in the art and are available through commercial suppliers.
- the antibodies of this disclosure may be produced recombinantly not only directly, but also as a fusion polypeptide with a heterologous polypeptide, which is preferably a signal sequence or other polypeptide having a specific cleavage site at the N-terminus of the mature protein or polypeptide.
- the homologous or heterologous signal sequence selected preferably is one that is recognized and processed (i.e., cleaved by a signal peptidase) by the host cell.
- mammalian signal sequences as well as viral secretory leaders, for example, the herpes simplex gD signal, are available.
- Such expression vectors and cloning vectors will generally contain a nucleic acid sequence that enables the vector to replicate in one or more selected host cells. Typically, in cloning vectors this sequence is one that enables the vector to replicate independently of the host chromosomal DNA, and includes origins of replication or autonomously replicating sequences. Such sequences are well known for a variety of bacteria, yeast, and viruses, e.g.
- the origin of replication from the plasmid pBR322 is suitable for most Gram-negative bacteria, the 2mu plasmid origin is suitable for yeast, and various viral origins (Simian Virus 40 (“SV40”), polyoma, adenovirus, vesicular stomatitis virus (“VSV”), or bovine papillomavirus (“BPV”) are useful for cloning vectors in mammalian cells.
- SV40 Sesimian Virus 40
- polyoma polyoma
- adenovirus vesicular stomatitis virus
- VSV vesicular stomatitis virus
- BBV bovine papillomavirus
- the origin of replication component is not needed for mammalian expression vectors (the SV40 origin may typically be used only because it contains the early promoter).
- These vectors will also typically contain a selection gene, also termed a selectable marker.
- Typical selection genes encode proteins that (a) confer resistance to antibiotics or other toxins, e.g., ampicillin, neomycin, methotrexate, or tetracycline, (b) complement auxotrophic deficiencies, or (c) supply critical nutrients not available from complex media, e.g., the gene encoding D-alanine racemase for Bacilli.
- One example of a selection scheme utilizes a drug to arrest growth of a host cell.
- Drug selection is generally used to select for cultured mammalian cells into which foreign DNA has been inserted. Such cells are commonly referred to as "transfectants”. Cells that have been cultured in the presence of the selective agent and are able to pass the gene of interest to their progeny are referred to as "stable transfectants.” Examples of such dominant selection use the drugs neomycin, mycophenolic acid, and hygromycin.
- An exemplary selectable marker is a gene encoding resistance to the antibiotic neomycin. Selection is carried out in the presence of a neomycin-type drug, such as G-418 or the like. Those cells that are successfully transformed with a heterologous gene produce a protein conferring drug resistance and thus survive the selection regimen.
- Selection systems can also be used to increase the expression level of the gene of interest, a process referred to as "amplification.”
- Amplification of transfectants typically occurs by culturing the cells in the presence of a low level of the selective agent and then increasing the amount of selective agent to select for cells that produce high levels of the products of the introduced genes.
- Suitable selectable markers for mammalian cells are those that enable the identification of cells competent to take up the antibody nucleic acid, such as dihydrofolate reductase (“DHFR”), thymidine kinase, metallothionein-I and -II, preferably primate metallothionein genes, adenosine deaminase, ornithine decarboxylase, etc.
- DHFR dihydrofolate reductase
- thymidine kinase thymidine kinase
- metallothionein-I and -II preferably primate metallothionein genes
- adenosine deaminase adenosine deaminase
- ornithine decarboxylase etc.
- an amplifiable selectable marker for mammalian cells is dihydrofolate reductase, which confers resistance to methotrexate.
- Other drug resistance genes e.g. hygromycin resistance, multi-drug resistance, puromycin acetyltransferase
- MTX methotrexate
- An appropriate host cell when wild-type DHFR is employed is the Chinese hamster ovary (“CHO”) cell line deficient in DHFR activity.
- host cells transformed or co-transformed with DNA sequences encoding antibody, wild-type DHFR protein, and another selectable marker such as aminoglycoside 3'-phosphotransferase (“APH”) can be selected by cell growth in medium containing a selection agent for the selectable marker such as an aminoglycosidic antibiotic, e.g., kanamycin, neomycin, or G-418. See U.S. Patent No. 4,965,199.
- APH aminoglycoside 3'-phosphotransferase
- These vectors may comprise an enhancer sequence that facilitates transcription of a DNA encoding the antibody.
- enhancer sequences are known from mammalian genes (for example, globin, elastase, albumin, alpha-fetoprotein, and insulin).
- a frequently used enhancer is one derived from a eukaryotic cell virus. Examples thereof include the SV40 enhancer on the late side of the replication origin (bp 100-270), the cytomegalovirus early promoter enhancer, the polyoma enhancer on the late side of the replication origin, and adenovirus enhancers (See also Yaniv, Nature, 297:17- 18 (1982) on enhancing elements for activation of eukaryotic promoters).
- the enhancer may be spliced into the vector at a position 5' or 3' to the antibody-encoding sequence, but is preferably located at a site 5' from the promoter.
- Expression and cloning vectors will also generally comprise a promoter that is recognized by the host organism and is operably linked to the antibody nucleic acid.
- Promoter sequences are known for eukaryotes. Virtually all eukaryotic genes have an AT-rich region located approximately 25 to 30 bases upstream from the site where transcription is initiated. Another sequence found 70 to 80 bases upstream from the start of transcription of many genes is a CNCAAT region where N may be any nucleotide. At the 3' end of most eukaryotic genes is an AATAAA sequence that may be the signal for addition of the poly A tail to the 3' end of the coding sequence. All of these sequences are suitably inserted into eukaryotic expression vectors.
- Antibody transcription from vectors in mammalian host cells may be controlled, for example, by promoters obtained from the genomes of viruses such as polyoma virus, fowlpox virus, adenovirus (such as Adenovirus 2), BPV, avian sarcoma virus, cytomegalovirus, a retrovirus, hepatitis-B virus, and most preferably SV40, from heterologous mammalian promoters, e.g., the actin promoter or an immunoglobulin promoter, from heat-shock promoters, provided such promoters are compatible with the host cell systems.
- viruses such as polyoma virus, fowlpox virus, adenovirus (such as Adenovirus 2), BPV, avian sarcoma virus, cytomegalovirus, a retrovirus, hepatitis-B virus, and most preferably SV40
- heterologous mammalian promoters e.g., the actin
- the early and late promoters of the SV40 virus are conveniently obtained as an SV40 restriction fragment that also contains the SV40 viral origin of replication.
- the immediate early promoter of the human cytomegalovirus is conveniently obtained as a Hindlll E restriction fragment.
- a system for expressing DNA in mammalian hosts using the BPV as a vector is disclosed in U.S. Patent No. 4,419,446. A modification of this system is described in U.S. Patent No. 4,601,978. See also Reyes et ai, Nature, 297:598-601 (1982) on expression of human beta-interferon cDNA in mouse cells under the control of a thymidine kinase promoter from herpes simplex virus. Alternatively, the Rous sarcoma virus long terminal repeat can be used as the promoter.
- Strong transcription promoters can be used, such as promoters from SV40, cytomegalovirus, or myeloproliferative sarcoma virus. See, e.g., U.S. Patent No. 4,956,288 and U.S. Patent Publication No. 20030103986.
- Other suitable promoters include those from metallothionein genes (U.S. Patent Nos. 4,579,821 and 4,601,978) and the adenovirus major late promoter.
- Expression vectors for use in mammalian cells include pZP-1, pZP-9, and pZMP21, which have been deposited with the American Type Culture Collection, 10801 University Boulevard., Manassas, VA. USA under accession numbers 98669, 98668, and PTA-5266, respectively, and derivatives of these vectors.
- Expression vectors used in eukaryotic host cells will also generally contain sequences necessary for the termination of transcription and for stabilizing the rnRNA. Such sequences are commonly available from the 5' and, occasionally 3', untranslated regions of eukaryotic or viral DNAs or cDNAs. These regions contain nucleotide segments transcribed as polyadenylated fragments in the untranslated portion of the rnRNA encoding the antibody.
- One useful transcription termination component is the bovine growth hormone polyadenylation region. See WO 94/11026 and the expression vector disclosed therein.
- Suitable host cells for cloning or expressing the subject antibodies include prokaryote, yeast, or higher eukaryote cells described above.
- interest has been greatest in vertebrate cells, and propagation of vertebrate cells in culture has become a routine procedure.
- useful mammalian host cell lines are monkey kidney CV1 line transformed by SV40 (COS-1 (ATCC No. CRL 1650); and COS-7, ATCC CRL 1651); human embryonic kidney line (293 or 293 cells subcloned for growth in suspension culture, (ATCC No. CRL 1573; Graham et ai, J. Gen.
- monkey kidney cells (CV1 ATCC CCL 70); African green monkey kidney cells (VERO-76, ATCC CRL-1587); human cervical carcinoma cells (HELA, ATCC CCL 2); canine kidney cells (MDCK, ATCC CCL 34); buffalo rat liver cells (BRL 3A, ATCC CRL 1442); human lung cells (W138, ATCC CCL 75); human liver cells (Hep G2, HB 8065); mouse mammary tumor (MMT 060562, ATCC CCL51); TRI cells (Mather et al, Annals N.Y. Acad.
- MRC 5 cells LS4 cells
- a human hepatoma line Hep G2
- Additional suitable cell lines are known in the art and available from public depositories such as the American Type Culture Collection, Manassas, VA.
- Host cells are transformed with the above -described expression or cloning vectors for antibody production and cultured in conventional nutrient media modified as appropriate for inducing promoters, selecting transformants, or amplifying the genes encoding the desired sequences as discussed supra.
- the mammalian host cells used to produce the antibody of this disclosure may be cultured in a variety of media.
- Commercially available media such as Ham's L10 (Sigma-Aldrich Corporation, St. Louis, MO), Minimal Essential Medium ((“MEM” (Sigma-Aldrich Corporation, St. Louis, MO), Roswell Park Memorial Institute- 1640 medium (“RPMI-1640”, Sigma-Aldrich Corporation, St.
- any of the media described in Ham et al, Meth. Enz ⁇ , 58:44 (1979), Barnes et aI., AhaI. Biochem., 102:255 (1980), U.S. Patent Nos. 4,767,704; 4,657,866; 4,927,762; 4,560,655; or 5,122,469; WO 90/03430; WO 87/00195; or U.S. Patent Reexam No. 30,985 can be used as culture media for the host cells.
- any of these media may be supplemented as necessary with hormones and/or other growth factors (such as insulin, transferrin, or epidermal growth factor), salts (such as sodium chloride, calcium, magnesium, and phosphate), buffers (such as HEPES), nucleotides (such as adenosine and thymidine), antibiotics (such as Gentamycin drug), trace elements (defined as inorganic compounds usually present at final concentrations in the micromolar range), and glucose or an equivalent energy source. Any other necessary supplements may also be included at appropriate concentrations that would be known to those skilled in the art.
- the culture conditions such as temperature, pH, and the like, are those previously used with the host cell selected for expression, and will be apparent to the ordinarily skilled artisan. Methods of development and optimization of media and culture conditions are known in the art (See, Gronemeyer et al., Bioengineering, 1(4): 188-212, 2014).
- these cells are cultured (either adherent cells or suspension cultures) most typically in a batch-fed process in a bioreactor (many models are commercially available) that involves continuously feeding the cell culture with medium and feed, optimized for the particular cell line chosen and selected for this purpose.
- a bioreactor many models are commercially available
- Perfusion systems are also available in which media and feed are continuously supplied to the culture while the same volume of media is being withdrawn from the bioreactor. (Wurm, 2004).
- Synthetic media also commercially available, are available for growing cells in a batch-fed culture, avoiding the possibility of contamination from outside sources, such as with the use of animal components, such as bovine serum albumin, etc.
- animal-component-free hydrolysates are commercially available to help boost cell density, culture viability and productivity. (Li et al, 2010). Many studies have been performed in an effort to optimize cell culture media, including careful attention to head space available in roller bottles, redox potentials during growth and expression phases, presence of reducing agents to maintain disulfide bonds during production, etc.
- Cultured cells may be grown by feeding nutrients continuously or as separately administered amounts. Often various process parameters such as cell concentration, pH, temperature, CO2, d0 2 , osmolality, amount of metabolites such as glucose, lactate, glutamine and glutamate, and the like, are monitored by the use of probes during the cell growth either on-line by direct connection to calibrated analyzers or off-line by intervention of operators.
- the culturing step also typically involves ensuring that the cells growing in culture maintain the transfected recombinant genes by any means known in the art for cell selection.
- the culturing step is typically followed by a harvesting step, whereby the cells are separated from the medium and a harvested cell culture media is thereby obtained.
- a harvesting step whereby the cells are separated from the medium and a harvested cell culture media is thereby obtained.
- various purification steps involving column chromatography and the like, follow culturing to separate the recombinant monoclonal antibody from cell components and cell culture media components.
- Centrifugation of cell components may be achieved on a large scale by use of continuous disk stack centrifuges followed by clarification using depth and membrane filters. (See, Kelley, 2009). Most often, after clarification, the recombinant protein is further purified by Protein A chromatography due to the high affinity of Protein A for the Fc domain of antibodies, and typically occurs using a low pH/acidification elution step (typically the acidification step is combined with a precautionary virus inactivation step). Flocculation and/or precipitation steps using acidic or cationic polyelectrolytes may also be employed to separate animal cells in suspension cultures from soluble proteins. (Liu et al, 2010).
- anion- and cation-exchange chromatography hydrophobic interaction chromatograph (“HIC”), hydrophobic charge induction chromatograph (HCIC), hydroxyapatite chromatography using ceramic hydroxyapatite (Cas(P04)30H)2, and combinations of these techniques are typically used to polish the solution of recombinant monoclonal antibody.
- Final formulation and concentration of the desired monoclonal antibody may be achieved by use of ultracentrifugation techniques. Purification yields are typically 70 to 80%. (Kelley, 2009).
- the terms “desired protein” or “desired antibody” herein are used interchangeably and refer generally to a parent antibody specific to a target, i.e., CoV-S or a chimeric or humanized antibody or a binding portion thereof derived therefrom as described herein.
- the term “antibody” is intended to include any polypeptide chain-containing molecular structure with a specific shape that fits to and recognizes an epitope, where one or more non-covalent binding interactions stabilize the complex between the molecular structure and the epitope.
- the archetypal antibody molecule is the immunoglobulin, and ah types of immunoglobulins, IgG, IgM, IgA, IgE, IgD, etc., from ah sources, e.g. human, rodent, rabbit, cow, sheep, pig, dog, other mammals, chicken, other avians, etc., are considered to be “antibodies.”
- ah sources e.g. human, rodent, rabbit, cow, sheep, pig, dog, other mammals, chicken, other avians, etc.
- examples thereof include chimeric antibodies, human antibodies and other non-human mammalian antibodies, humanized antibodies, single chain antibodies (such as scFvs), camelbodies, nanobodies, IgNAR (single-chain antibodies which may be derived from sharks, for example), small-modular immunopharmaceuticals (“SMIPs”), and antibody fragments such as Fabs, Fab', F(ab')2, and the like ( See Strel
- antibodies or antigen-binding fragments thereof may be produced by genetic engineering.
- antibody-producing cells are sensitized to the desired antigen or immunogen.
- the messenger RNA isolated from antibody producing cells is used as a template to make cDNA using PCR amplification.
- a library of vectors, each containing one heavy chain gene and one light chain gene retaining the initial antigen specificity, is produced by insertion of appropriate sections of the amplified immunoglobulin cDNA into the expression vectors.
- a combinatorial library is constructed by combining the heavy chain gene library with the light chain gene library.
- Antibody coding sequences of interest include those encoded by native sequences, as well as nucleic acids that, by virtue of the degeneracy of the genetic code, are not identical in sequence to the disclosed nucleic acids, and variants thereof.
- Variant polypeptides can include amino acid (“aa”) substitutions, additions, or deletions.
- amino acid substitutions can be conservative amino acid substitutions or substitutions to eliminate non-essential amino acids, such as to alter a glycosylation site, or to minimize misfolding by substitution or deletion of one or more cysteine residues that are not necessary for function.
- Variants can be designed so as to retain or have enhanced biological activity of a particular region of the protein (e.g., a functional domain, catalytic amino acid residues, etc).
- Variants also include fragments of the polypeptides disclosed herein, particularly biologically active fragments and/or fragments corresponding to functional domains. Techniques for in vitro mutagenesis of cloned genes are known. Also included in the subject disclosure are polypeptides that have been modified using ordinary molecular biological techniques so as to improve their resistance to proteolytic degradation or to optimize solubility properties or to render them more suitable as a therapeutic agent.
- Chimeric antibodies may be made by recombinant means by combining the V L and V H regions, obtained from antibody producing cells of one species with the constant light and heavy chain regions from another.
- chimeric antibodies utilize rodent or rabbit variable regions and human constant regions, in order to produce an antibody with predominantly human domains.
- chimeric antibodies The production of such chimeric antibodies is well known in the art, and may be achieved by standard means (as described, e.g., in U.S. Patent No. 5,624,659, incorporated herein by reference in its entirety). It is further contemplated that the human constant regions of chimeric antibodies of the disclosure may be selected from IgGl, IgG2, IgG3, and IgG4 constant regions.
- Humanized antibodies are engineered to contain even more human-like mmunoglobulin domains, and incorporate only the complementarity determining regions of the animal -derived antibody. This is accomplished by carefully examining the sequence of the hyper- variable loops of the variable regions of the monoclonal antibody and fitting them to the structure of the human antibody chains. Although facially complex, the process is straightforward in practice. See, e.g., U.S. Patent No. 6,187,287, incorporated fully herein by reference.
- immunoglobulin fragments comprising the epitope binding site (e.g., Fab’, F(ab’)2, or other fragments) may be synthesized.
- “Fragment” or minimal immunoglobulins may be designed utilizing recombinant immunoglobulin techniques.
- Fv immunoglobulins for use in the present disclosure may be produced by synthesizing a fused variable light chain region and a variable heavy chain region. Combinations of antibodies are also of interest, e.g. diabodies, which comprise two distinct Fv specificities.
- SMIPs small molecule immunopharmaceuticals
- camelbodies camelbodies
- nanobodies and IgNAR are encompassed by immunoglobulin fragments.
- Immunoglobulins and fragments thereof may be modified post-translationally, e.g. to add effector moieties such as chemical linkers, detectable moieties, such as fluorescent dyes, enzymes, toxins, substrates, bioluminescent materials, radioactive materials, chemiluminescent moieties, and the like, or specific binding moieties, such as streptavidin, avidin, or biotin, and the like may be utilized in the methods and compositions of the present disclosure. Examples of additional effector molecules are provided infra.
- a "heterologous" region or domain of a DNA construct is an identifiable segment of DNA within a larger DNA molecule that is not found in association with the larger molecule in nature.
- the DNA flanking the gene usually does not flank the mammalian genomic DNA in the genome of the source organism.
- a heterologous region is a construct where the coding sequence itself is not found in nature (e.g., a cDNA where the genomic coding sequence contains introns or synthetic sequences having codons different than the native gene). Allelic variations or naturally-occurring mutational events do not give rise to a heterologous region of DNA as defined herein.
- a "coding sequence” is an in-frame sequence of codons that correspond to or encode a protein or peptide sequence. Two coding sequences correspond to each other if the sequences or their complementary sequences encode the same amino acid sequences. A coding sequence in association with appropriate regulatory sequences may be transcribed and translated into a polypeptide. A polyadenylation signal and transcription termination sequence will usually be located 3' to the coding sequence.
- a "promoter sequence” is a DNA regulatory region capable of initiating transcription of a downstream (3' direction) coding sequence, and typically contain additional sites for binding of regulatory molecules, e.g., transcription factors, that affect the transcription of the coding sequence.
- a coding sequence is "under the control" of the promoter sequence or "operatively linked” to the promoter when RNA polymerase binds the promoter sequence in a cell and transcribes the coding sequence into mRNA, which is then in turn translated into the protein encoded by the coding sequence.
- Antibodies consist of two identical light polypeptide chains of molecular weight approximately 23,000 daltons (the “light chain”), and two identical heavy chains of molecular weight 53,000-70,000 (the “heavy chain”). The four chains are joined by disulfide bonds in a “Y” configuration wherein the light chains bracket the heavy chains starting at the mouth of the “Y” configuration.
- the “branch” portion of the “Y” configuration is designated the F ab region; the stem portion of the “Y” configuration is designated the Fc region.
- the amino acid sequence orientation runs from the N-terminal end at the top of the “Y” configuration to the C-terminal end at the bottom of each chain.
- the N-terminal end possesses the variable region having specificity for the antigen that elicited it, and is approximately 100 amino acids in length, there being slight variations between light and heavy chain and from antibody to antibody.
- variable region is linked in each chain to a constant region that extends the remaining length of the chain and that within a particular class of antibody does not vary with the specificity of the antibody (i.e., the antigen eliciting it).
- constant regions There are five known major classes of constant regions that determine the class of the immunoglobulin molecule (IgG, IgM, IgA, IgD, and IgE corresponding to g, m, a, d, and e (gamma, mu, alpha, delta, or epsilon) heavy chain constant regions).
- the constant region or class determines subsequent effector function of the antibody, including activation of complement ( see Kabat, E.
- variable region refers to the domains within each pair of light and heavy chains in an antibody that are involved directly in binding the antibody to the antigen.
- Each heavy chain has at one end a variable region (VFI) followed by a number of constant domains.
- Each light chain has a variable region (VL) at one end and a constant domain at its other end; the constant domain of the light chain is aligned with the first constant domain of the heavy chain, and the light chain variable domain is aligned with the variable domain of the heavy chain.
- CDR complementarity-determining region
- hypervariable region refers to one or more of the hyper-variable or complementarity-determining regions (“CDRs”) found in the variable regions of light or heavy chains of an antibody ( See Kabat et ai, Sequences of Proteins of Immunological Interest, 4 th ed., Bethesda, MD: U.S. Dept of Flealth and Human Services, Public Health Service, National Institutes of Health (1987)). These expressions include the hypervariable regions as defined by Kabat et ai, ( Sequences of Proteins of Immunological Interest, NIH Publication No. 91-3242, Bethesda, MD: U.S. Dept of Health and Human Services, National Institutes of Health (1983)) or the hypervariable loops in 3-dimensional structures of antibodies (Chothia and Lesk, J.
- the CDRs in each chain are held in close proximity by framework regions (“FRs”) and, with the CDRs from the other chain, contribute to the formation of the antigen binding site.
- FRs framework regions
- select amino acids that have been described as the selectivity determining regions (“SDRs”) that represent the critical contact residues used by the CDR in the antibody-antigen interaction ( see Kashmiri et al, Methods, 36(l):25-34 (2005)).
- An "epitope" or “binding site” is an area or region on an antigen to which an antigen-binding peptide (such as an antibody) specifically binds.
- a protein epitope may comprise amino acid residues directly involved in the binding (also called immunodominant component of the epitope) and other amino acid residues, which are not directly involved in the binding, such as amino acid residues that are effectively blocked by the specifically antigen binding peptide (in other words, the amino acid residue is within the "footprint" of the specifically antigen binding peptide).
- epitope herein includes both types of amino acid binding sites in any particular region of CoV-S, e.g., SARS-CoV-S or SARS-CoV-2-S, that specifically binds to an anti-CoV-S antibody.
- CoV-S may comprise a number of different epitopes, which may include, without limitation, (1) linear peptide antigenic determinants, (2) conformational antigenic determinants that consist of one or more non-contiguous amino acids located near each other in a mature CoV-S conformation; and (3) post-translational antigenic determinants that consist, either in whole or part, of molecular structures covalently attached to a CoV-S protein such as carbohydrate groups.
- epitope includes the specific residues in a protein or peptide, e.g., CoV-S, which are involved in the binding of an antibody to such protein or peptide as determined by known and accepted methods such as alanine scanning techniques or the use of various S protein portions with varying lengths.
- an antibody binds "substantially" or “at least partially” the same epitope as another antibody (e.g., second antibody) means that the epitope binding site for the first antibody comprises at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more of the amino acid residues on the antigen that constitutes the epitope binding site of the second antibody.
- a first antibody binds substantially or partially the same or overlapping epitope as a second antibody means that the first and second antibodies compete in binding to the antigen, as described above.
- the term "binds to substantially the same epitope or determinant as" a monoclonal antibody means that an antibody "competes" with the antibody.
- the phrase "binds to the same or overlapping epitope or determinant as" an antibody of interest means that an antibody "competes" with said antibody of interest for at least one, (e.g., at least 2, at least 3, at least 4, at least 5) or all residues on CoV-S to which said antibody of interest specifically binds.
- the identification of one or more antibodies that bind(s) to substantially or essentially the same epitope as the monoclonal antibodies described herein can be readily determined using alanine scanning. Additionally, any one of variety of immunological screening assays in which antibody competition can be assessed. A number of such assays are routinely practiced and well known in the art (see, e.g., U.S. Patent No.
- test antibodies to be examined are obtained from different source animals, or are even of a different Ig isotype, a simple competition assay may be employed in which the control antibody is mixed with the test antibody and then applied to a sample containing CoV-S. Protocols based upon ELISAs, radioimmunoassays, Western blotting, and the use of BIACORE®
- control anti-CoV-S antibody is pre-mixed with varying amounts of the test antibody (e.g., in ratios of about 1:1, 1:2, 1:10, or about 1:100) for a period of time prior to applying to the CoV-S (e.g., SARS-CoV-S or SARS-CoV-2-S) antigen sample.
- the control and varying amounts of test antibody can simply be added separately and admixed during exposure to the SARS-CoV-S or SARS-CoV-2-S antigen sample.
- bound antibodies can be distinguished from free antibodies (e.g., by using separation or washing techniques to eliminate unbound antibodies) and control antibody from the test antibody (e.g., by using species specific or isotype specific secondary antibodies or by specifically labeling the control antibody with a detectable label) it can be determined if the test antibody reduces the binding of the control antibody to the SARS-CoV-S or SARS-CoV-2-S antigens, indicating that the test antibody recognizes substantially the same epitope as the control anti-CoV-S antibody.
- the binding of the (labeled) control antibody in the presence of a completely irrelevant antibody (that does not bind CoV-S) can serve as the control high value.
- the control low value can be obtained by incubating the labeled control antibody with the same but unlabeled control antibody, where competition would occur and reduce binding of the labeled antibody.
- a significant reduction in labeled antibody reactivity in the presence of a test antibody is indicative of a test antibody that recognizes substantially the same epitope, i.e., one that competes with the labeled control antibody.
- any test antibody that reduces the binding of the control antibody to SARS-CoV-S or SARS-CoV-2-S by at least about 50%, such as at least about 60%, or more preferably at least about 70% (e.g., about 65-100%), at any ratio of test antibody between about 1:1 or 1:10 and about 1:100 is considered to be an antibody that binds to substantially the same or overlapping epitope or determinant as the control antibody.
- test antibody will reduce the binding of the control antibody to SARS-CoV-S or SARS-CoV-2-S (or another CoV-S) antigen preferably at least about 50%, at least about 60%, at least about 80%, or at least about 90% (e.g., about 95%) of the binding of the control antibody observed in the absence of the test antibody.
- a simple competition assay in which a test antibody is applied at saturating concentration to a surface onto which SARS-CoV-S or SARS-CoV-2-S (or another CoV-S) is immobilized also may be advantageously employed.
- the surface in the simple competition assay is preferably a BIACORE® (GE Healthcare Life Sciences, Marlborough, MA) chip (or other media suitable for surface plasmon resonance (“SPR”) analysis).
- SPR surface plasmon resonance
- This binding to the SARS-CoV-S- or SARS- CoV -2 -S -containing surface of the control antibody alone is compared with the binding of the control antibody in the presence of a test antibody.
- a significant reduction in binding to the SARS-CoV-S- or S ARS -Co V -2 -S -containing surface by the control antibody in the presence of a test antibody indicates that the test antibody recognizes substantially the same epitope as the control antibody such that the test antibody "competes" with the control antibody.
- Any test antibody that reduces the binding of control antibody by at least about 20% or more, at least about 40%, at least about 50%, at least about 70%, or more, can be considered to be an antibody that binds to substantially the same epitope or determinant as the control antibody.
- test antibody will reduce the binding of the control antibody to SARS-CoV-S or SARS-CoV-2-S by at least about 50% (e.g., at least about 60%, at least about 70%, or more).
- control and test antibodies can be reversed; i.e. the control antibody can be first bound to the surface and then the test antibody is brought into contact with the surface thereafter in a competition assay.
- the “sandwich- style” binding assay infra is used.
- the antibody having greater affinity for SARS-CoV-S or SARS-CoV-2-S antigen is bound to the SARS-CoV-S- or SARS-CoV-2-S-containing surface first, as it will be expected that the decrease in binding seen for the second antibody (assuming the antibodies are competing) will be of greater magnitude.
- assays are provided in e.g., Saunal and Regenmortel, J. Immunol. Methods, 183:33-41 (1995), the disclosure of which is incorporated herein by reference.
- an antibody binds the same or overlapping epitope(s) on COV-S as another antibody or the epitope bound by a test antibody may in particular be determined using a Western-blot based assay.
- a library of peptides corresponding to the antigen bound by the antibody, the CoV-S protein is made, that comprise overlapping portions of the protein, typically 10- 25, 10-20, or 10-15 amino acids long.
- These different overlapping amino acid peptides encompassing the CoV-S sequence are synthesized and covalently bound to a PEPSPOTSTM nitrocellulose membrane (JPT Peptide Technologies, Berlin, Germany). Blots are then prepared and probed according to the manufacturer's recommendations.
- the immunoblot assay detects by fluorometric means what peptides in the library bind to the test antibody and thereby can identify what residues on the antigen, i.e., COV-S, interact with the test antibody. (See U.S. Patent No. 7,935,340, incorporated by reference herein).
- fluorometric means what peptides in the library bind to the test antibody and thereby can identify what residues on the antigen, i.e., COV-S, interact with the test antibody.
- X-ray co- crystallography of the antigen and antibody NMR; SPR (e.g., at 25° or 37°C); array-based oligo peptide scanning (or “pepscan analysis”); site -directed mutagenesis (e.g., alanine scanning); mutagenesis mapping; hydrogen-deuterium exchange; phage display; and limited proteolysis are all epitope mapping techniques that are well known in the art (See, e.g., Epitope Mapping Protocols: Second Edition, Methods in Molecular Biology, , editors Mike Schutkowski and Ulrich Reineke, 2 nd Ed., New York, NY: Humana Press (2009), and Epitope Mapping Protocols, Methods in Molecular Biology, editor Glenn Morris, 1 st Ed., New York, NY: Humana Press (1996), both of which are herein incorporated by referenced in their entirety).
- any one of antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, ADI-58131 can be readily determined using any one of variety of immunological screening assays in which antibody competition can be assessed. A number of such assays are routinely practiced and well known in the art (see, e.g., U.S. Patent No.
- test antibodies to be examined are obtained from different source animals, or are even of a different Ig isotype
- a simple competition assay may be employed in which the control antibody (one of antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, ADI-58131, for example) is mixed with the test antibody and then applied to a sample containing either or both SARS-CoV-S or SARS-CoV-2-S, each of which is known to be bound by antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, ADI-
- Protocols based upon ELISAs, radioimmunoassays, Western blotting, and BIACORE® (GE Healthcare Life Sciences, Marlborough, MA) analysis are suitable for use in such simple competition studies.
- the method comprises pre-mixing the control antibody with varying amounts of the test antibody (e.g., in ratios of about 1:1, 1:2, 1:10, or about 1:100) for a period of time prior to applying to the CoV-S antigen sample.
- the control and varying amounts of test antibody can be added separately and admixed during exposure to the CoV-S antigen sample.
- the method can be used to determine that the test antibody reduces the binding of the control antibody to the COV-S antigen, indicating that the test antibody recognizes substantially the same epitope as the control antibody (e.g., antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI- 58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, ADI-58131).
- the binding of the (labeled) control antibody in the presence of a completely irrelevant antibody (that does not bind CoV-S) can serve as the control high value.
- the control low value can be obtained by incubating the labeled control antibody with the same but unlabeled control antibody, where competition would occur and reduce binding of the labeled antibody.
- a significant reduction in labeled antibody reactivity in the presence of a test antibody is indicative of a test antibody that recognizes substantially the same epitope, i.e., one that competes with the labeled control antibody.
- any test antibody that reduces the binding of any one of antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI- 58127, ADI-58128, ADI-58129, ADI-58130, ADI-58131, to both of SARS-CoV-S or SARS-CoV-2-S antigens by at least about 50%, such as at least about 60%, or more preferably at least about 70%
- control antibody selected from the group consisting of ADI- 58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI- 58128, ADI-58129, ADI-58130, ADI-58131
- test antibody between about 1:1 or 1:10 and about 1:100 is considered to be an antibody that binds to substantially the same epitope or determinant as any one of antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, ADI-58131, respectively.
- such test antibody will reduce the binding of any one of antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI- 58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, ADI-58131, to at least one, preferably each, of the SARS-CoV-S or SARS-CoV-2-S antigens preferably at least about 50%, at least about 60%, at least about 80% or at least about 90% (e.g., about 95%) of the binding of any one of antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, ADI-58131, observed in the absence of the test antibody.
- a simple competition assay in which a test antibody is applied at saturating concentration to a surface onto which either SARS-CoV-S or SARS-CoV-2-S, or both, are immobilized also may be advantageously employed.
- the surface in the simple competition assay is preferably of a media suitable for OCTET® and/or PROTEON®.
- control antibody e.g., any one of antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, ADI-58131
- This binding to the CoV-S-containing surface of the control antibody alone is compared with the binding of the control antibody in the presence of a test antibody.
- a significant reduction in binding to the CoV-S-containing surface by the control antibody in the presence of a test antibody indicates that the test antibody recognizes substantially the same epitope as the control antibody such that the test antibody "competes" with the control antibody.
- Any test antibody that reduces the binding of control antibody (such as anyone of antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI- 58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, ADI-58131) to both of SARS-CoV-S and SARS-CoV-2-S antigens by at least about 20% or more, at least about 40%, at least about 50%, at least about 70%, or more, can be considered to be an antibody that binds to substantially the same epitope or determinant as the control antibody (e.g., any one of antibodies selected from the group consisting of ADI-58120,
- such test antibody will reduce the binding of the control antibody (e.g., any one of antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, ADI-58131) to the CoV-S antigen by at least about 50% (e.g., at least about 60%, at least about 70%, or more).
- the order of control and test antibodies can be reversed; i.e. the control antibody can be first bound to the surface and then the test antibody is brought into contact with the surface thereafter in a competition assay.
- the antibody having higher affinity for SARS-CoV-S and SARS-CoV-2-S is bound to the CoV-S-containing surface first, as it will be expected that the decrease in binding seen for the second antibody (assuming the antibodies are competing) will be of greater magnitude.
- assays are provided in, e.g., Saunal and Regenmortel, J. Immunol. Methods, 183:33-41 (1989), the disclosure of which is incorporated herein by reference.
- an epitope region for an anti-CoV-S antibody may be determined by epitope "footprinting" using chemical modification of the exposed amines/carboxyls in the SARS- CoV-S and SARS-CoV-2-S protein.
- HXMS hydrogen-deuterium exchange detected by mass spectrometry
- a hydrogen/deuterium exchange of receptor and ligand protein amide protons, binding, and back exchange occurs, wherein the backbone amide groups participating in protein binding are protected from back exchange and therefore will remain deuterated.
- Relevant regions can be identified at this point by peptic proteolysis, fast microbore high-performance liquid chromatography separation, and/or electrospray ionization mass spectrometry (See, e.g., Ehring H., Analytical Biochemistry, 267(2):252-259 (1999) and Engen, J. R. & Smith, D. L., Anal.
- NMR nuclear magnetic resonance epitope mapping
- the antigen typically is selectively isotopically labeled with 15 N so that only signals corresponding to the antigen and no signals from the antigen binding peptide are seen in the NMR- spectrum.
- Antigen signals originating from amino acids involved in the interaction with the antigen binding peptide typically will shift position in the spectras of the complex compared to the spectras of the free antigen, and the amino acids involved in the binding can be identified that way. See, e.g., Ernst Sobering Res. Found. Workshop, (44):149-67 (2004); Huang etal., J. Mol. Biol., 281(1):61 -67 (1998); and Saito and Patterson, Methods, 9(3):516-24 (1996). Epitope mapping/characterization also can be performed using mass spectrometry (“MS”) methods ⁇ See, e.g., Downard, J. Mass Spectrom., 35(4):493-503 (2000) and Kiselar and Downard, Anal. Chem., 71(9):1792-801 (1999)).
- MS mass spectrometry
- Protease digestion techniques also can be useful in the context of epitope mapping and identification.
- Antigenic determinant-relevant regions/sequences can be determined by protease digestion, e.g. by using trypsin in a ratio of about 1:50 to SARS-CoV-S or SARS-CoV-2-S overnight (“o/n”) digestion at 37°C and pH 7-8, followed by mass spectrometry (“MS”) analysis for peptide identification.
- MS mass spectrometry
- the peptides protected from trypsin cleavage by the anti-CoV-S antibody can subsequently be identified by comparison of samples subjected to trypsin digestion and samples incubated with antibody and then subjected to digestion by e.g.
- Site -directed mutagenesis is another technique useful for characterization of a binding epitope.
- site -directed mutagenesis also known as alanine scanning, alanine scanning mutagenesis, alanine scanning mutations, combinatorial alanine scanning, or creation of alanine point mutations, for example
- each residue within a protein segment is replaced with an alanine residue (or another residue such as valine where alanine is present in the wild-type sequence) through such methodologies as direct peptide or protein synthesis, site-directed mutagenesis, the GENEARTTM Mutagenesis Service (Thermo Fisher Scientific, Waltham, MA U.S.A.) or shotgun mutagenesis, for example.
- a series of single point mutants of the molecule is thereby generated using this technique; the number of mutants generated is equivalent to the number of residues in the molecule, each residue being replaced, one at a time, by a single alanine residue.
- Alanine is generally used to replace native (wild-type) residues because of its non-bulky, chemically inert, methyl functional group that can mimic the secondary structure preferences that many other amino acids may possess.
- the effects replacing a native residue with an alanine has on binding affinity of an alanine scanning mutant and its binding partner can be measured using such methods as, but not limited to, SPR binding experiments. If a mutation leads to a significant reduction in binding affinity, it is most likely that the mutated residue is involved in binding.
- Monoclonal antibodies specific for structural epitopes can be used as a positive control for binding affinity experiments to verify that the alanine -replacement does not influence the overall tertiary structure of the protein (as changes to the overall fold of the protein may indirectly affect binding and thereby produce a false positive result).
- alanine -replacement does not influence the overall tertiary structure of the protein (as changes to the overall fold of the protein may indirectly affect binding and thereby produce a false positive result).
- Clackson and Wells Science, 267:383-386 (1995); Weiss et al, Proc. Natl. Acad. Sci. USA, 97(16):8950-8954 (2000); and Wells, Proc. Natl. Acad. Sci. USA, 93:1-6 (1996).
- Example 5 identifies the specific epitope or residues of CoV-S which specifically interact with the anti-CoV-S antibodies disclosed herein.
- Electron microscopy can also be used for epitope "footprinting".
- Wang et al. Nature, 355:275-278 (1992) used coordinated application of cryoelectron microscopy, three- dimensional image reconstruction, and X-ray crystallography to determine the physical footprint of a Fab-fragment on the capsid surface of native cowpea mosaic virus.
- label-free assay for epitope evaluation include SPR (sold commercially as the BIACORE® system, GE Healthcare Life Sciences, Marlborough, MA) and reflectometric interference spectroscopy (“RifS”) (See, e.g., Fagerstam et al, Journal of Molecular Recognition, 3:208-14 (1990); Nice et al, J. Chromatogr., 646:159-168 (1993); Leipert et al, Angew. Chem. Int. Ed., 37:3308-3311 (1998); Kroger et al, Biosensors and Bioelectronics, 17:937-944 (2002)).
- framework region refers to one or more of the framework regions within the variable regions of the light and heavy chains of an antibody ( See Rabat et al. , Sequences of Proteins of Immunological Interest, 4 th edition, Bethesda, MD: U.S. Dept of Health and Human Services, Public Health Service, National Institutes of Health (1987)). These expressions include those amino acid sequence regions interposed between the CDRs within the variable regions of the light and heavy chains of an antibody.
- the term "Fc region” is used to define a C-terminal region of an immunoglobulin heavy chain.
- the "Fc region” may be a native sequence Fc region or a variant Fc region.
- the human IgG heavy chain Fc region is usually defined to stretch from an amino acid residue at position Cys226, or from Pro230, to the carboxyl-terminus thereof.
- the numbering of the residues in the Fc region is that of the EU index as in Rabat. Rabat et al, Sequences of Proteins of Immunological Interest, 5th edition, Bethesda, MD: U.S. Dept of Health and Human Services, Public Health Service, National Institutes of Health (1991).
- the Fc region of an immunoglobulin generally comprises two constant domains, Cm and CH3.
- Fc receptor and “FcR” describe a receptor that binds to the Fc region of an antibody.
- the preferred FcR is a native sequence human FcR.
- a preferred FcR is one that binds an IgG antibody (a gamma receptor) and includes receptors of the FcyRI, FcyRII, and FcyRIII subclasses, including allelic variants and alternatively spliced forms of these receptors.
- FcyRII receptors include FcyRIIA (an “activating receptor”) and FcyRIIB (an “inhibiting receptor”), which have similar amino acid sequences that differ primarily in the cytoplasmic domains thereof.
- FcRs are reviewed in Ravetch and Kinet, Ann. Rev. Immunol., 9:457-92 (1991); Capel et al, Immunomethods, 4:25-34 (1994); and de Haas et al, J. Lab. Clin. Med., 126:330-41 (1995).
- FcR also includes the neonatal receptor, FcRn, which is responsible for the transfer of maternal IgGs to the fetus (Guyer et al, J. Immunol., 117:587 (1976); and Kim et al, J. Immunol., 24:249 (1994)), and which primarily functions to modulate and/or extend the half-life of antibodies in circulation.
- the disclosed anti-CoV-S antibodies are aglycosylated, as a result of the expression system and/or sequence, the subject antibodies are expected to bind FcRn receptors, but not to bind (or to minimally bind) Fey receptors.
- a "functional Fc region” possesses at least one effector function of a native sequence Fc region.
- effector functions include Clq binding; complement dependent cytotoxicity (“CDC”); Fc receptor binding; antibody-dependent cell-mediated cytotoxicity (“ADCC”); phagocytosis; down-regulation of cell surface receptors (e.g. B cell receptor (“BCR”)), etc.
- Such effector functions generally require the Fc region to be combined with a binding domain (e.g. an antibody variable domain) and can be assessed using various assays known in the art for evaluating such antibody effector functions.
- a “native sequence Fc region” comprises an amino acid sequence identical to the amino acid sequence of an Fc region found in nature.
- a “variant Fc region” comprises an amino acid sequence that differs from that of a native sequence Fc region by virtue of at least one amino acid modification, yet retains at least one effector function of the native sequence Fc region.
- the variant Fc region has at least one amino acid substitution compared to a native sequence Fc region or to the Fc region of a parent polypeptide, e.g. from about one to about ten amino acid substitutions, and preferably from about one to about five amino acid substitutions in a native sequence Fc region or in the Fc region of the parent polypeptide.
- the variant Fc region herein will preferably possess at least about 80% sequence identity with a native sequence Fc region and/or with an Fc region of a parent polypeptide, and most preferably at least about 90% sequence identity therewith, more preferably at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% sequence identity therewith.
- the Fc region of an antibody or antigen-binding antibody fragment of the present disclosure may bind to an Fc receptor (FcR).
- the FcR may be, but is not limited to, Fc gamma receptor (FcgR), FcgRI, FcgRIIA, FcgRIIBl, FcgRIIB2, FcgRIIIA, FcgRIIIB, Fc epsilon receptor (FceR), FceRI, FceRII, Fc alpha receptor (FcaR), FcaRI, Fc alpha/mu receptor (Fca/mR), or neonatal Fc receptor (FcRn).
- the Fc may be an IgM, IgD, IgG, IgE, or IgA isotype.
- An IgG isotype may be an IgGl, IgG2, IgG3, or IgG4.
- Certain amino acid modifications in the Fc region are known to modulate Ab effector functions and properties, such as, but not limited to, antibody-dependent cellular cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP), complement dependent cytotoxicity (CDC), and half -life (Wang X. et ah, Protein Cell. 2018 Jan; 9(1): 63-73; DalFAcqua W. F. et ah, J Biol Chem. 2006 Aug 18;281(33):23514-24. Epub 2006 Jun 21; Monnet C. et al, Front Immunol. 2015 Feb 4;6:39. doi: 10.3389/fimmu.2015.00039. eCollection 2015).
- ADCC antibody-dependent cellular cytotoxicity
- ADCP antibody-dependent cellular phagocytosis
- CDC complement dependent cytotoxicity
- the mutation may be symmetrical or asymmetrical.
- antibodies with Fc regions that have asymmetrical mutation(s) may provide better functions such as ADCC (Fiu Z. et al. J Biol Chem. 2014 Feb 7; 289(6): 3571-3590).
- any of the antibody variable region sequences disclosed herein may be used in combination with a wild-type (WT) Fc or a variant Fc.
- WT wild-type
- an Fc selected from the Fc sequences described in Table 9 may be used.
- Any of the variable region sequences disclosed herein may be used in combination with any appropriate Fc including any of the Fc variants provided in Table 9 to form an antibody or an antigen-binding antibody fragment of the present disclosure.
- the lysine (K) at the C-terminus of each Fc may be present or absent.
- An IgGl -type Fc optionally may comprise one or more amino acid substitutions.
- substitutions may include, for example, N297A, N297Q, D265A, F234A, F235A, C226S, C229S, P238S, E233P, F234V, G236-deleted, P238A, A327Q, A327G, P329A, K322A, F234F, F235E, P331S, T394D, A330F, P331S, F243F, R292P, Y300F, V305I, P396F, S239D, I332E, S298A, E333A, K334A, F234Y, F235Q, G236W, S239M, H268D, D270E, K326D, A330M, K334E, G236A, K326W, S239D, E333S, S267
- the Fc region may further comprise one or more additional amino acid substitutions. Such substitutions may include but are not limited to A330F, F234F, F235E, P3318, and/or any combination thereof (the residue numbering is according to the EU index as in Rabat).
- substitution combinations for an IgGl -type Fc include, but not limited to: M252Y, S254T, and T256E (“YTE” variant); M428F and N434A (“FA” variant), M428F and N434S (“LS” variant); M428L, N434A, Q438R, and S440E (“LA-RE” variant); L432D and N434L (“DEL” variant); and L234A, L235A, L432D, and N434L (“LALA-DEL” variant) (the residue numbering is according to the EU index as in Rabat).
- an IgGl-type Fc variant may comprise the amino acid sequence of SEQ ID NOS: 411, 412, 413, 414, 415, 416, or 417.
- the Fc variant is an LA variant and comprises the amino acid sequence of SEQ ID NO: 413.
- the Fc region optionally may comprise one or more amino acid substitutions.
- substitutions may include but are not limited to P238S, V234A, G237A, H268A, H268Q, H268E, V309L, N297A, N297Q, A330S, P331S, C232S, C233S, M252Y, S254T, T256E, and/or any combination thereof (the residue numbering is according to the EU index as in Rabat).
- the Fc region optionally may further comprise one or more additional amino acid substitutions. Such substitutions may include but are not limited to M252Y, S254T, T256E, and/or any combination thereof (the residue numbering is according to the EU index as in Rabat).
- An IgG3-type Fc region optionally may comprise one or more amino acid substitutions.
- Such substitutions may include but are not limited to E235Y (the residue numbering is according to the EU index as in Rabat).
- An IgG4-type Fc region optionally may comprise one or more amino acid substitutions.
- Such substitutions may include but are not limited to, E233P, F234V, L235A, G237A, E318A, S228P, L236E, S241P, L248E, T394D, M252Y, S254T, T256E, N297A, N297Q, and/or any combination thereof (the residue numbering is according to the EU index as in Rabat).
- the substitution may be, for example, S228P (the residue numbering is according to the EU index as in Rabat).
- the glycan of the hum an -like Fc region may be engineered to modify the effector function (for example, see Li T. et al, Proc Natl Acad Sci USA. 2017 Mar 28; 114(13):3485- 3490. doi: 10.1073/pnas.l702173114. Epub 2017 Mar 13).
- an “isolated” antibody refers to an antibody that is substantially free of other antibodies having different antigenic specificities. In some embodiments, an isolated antibody is substantially free of other unintended cellular material and/or chemicals.
- telomere binding means that the interaction of the antibody, or antigen-binding portion thereof, with an antigen is dependent upon the presence of a particular structure (e.g., antigenic determinant or epitope).
- the antibody, or antigen binding portion thereof binds to a specific protein, rather than proteins generally.
- an antibody, or antigen-binding portion thereof specifically binds a target, e.g., SARS- CoV-S and/or SARS-CoV-S-2.
- an antibody, or antigen-binding portion thereof specifically binds to more than one coronavirus spike protein, e.g., the spike protein of SARS-CoV-S and the spike protein of SARS-CoV-2-S, for example.
- the antibody, or antigen-binding portion thereof specifically binds to two different, but related, antigens, e.g., the spike protein of SARS-CoVl-S and the spike protein of SARS-CoV2-S, e.g., via a conserved epitope.
- CoV-S refers to the S protein of a coronavirus which is expressed on the surface of virions as a structural protein. As mentioned previously, the S protein plays an essential role for coronaviruses in binding to receptors on the host cell and determines host tropism (Zhu Z. et al. , Infect Genet Evol. 2018 Jul;61 : 183-184).
- SARS-CoV and SARS-CoV-2 bind to angiotensin-converting enzyme 2 (ACE2) of the host cell via the S protein’s receptor-binding domains (RBDs) and uses ACE2 as a receptor to enter the host cells (Ge X.Y. et al, Nature. 2013 Nov 28;503(7477):535-8. doi:
- SARS-CoV can also use CD209L (also known as L-SIGN) as an alternative receptor (Jeffers S. A. et al, Proc Natl Acad Sci USA. 2004 Nov 2;101(44):15748-53. Epub 2004 Oct 20).
- MERS-CoV binds dipeptidyl peptidase 4 (“DPP4”, also known as CD26) of the host cells via a different RBD of the S protein.
- DPP4 dipeptidyl peptidase 4
- the S protein of SARS-CoV is referred to as SARS-CoV-S and may for example comprise the amino acid sequence of SEQ ID NO: 401 (1288 amino acids).
- the S protein of SARS-CoV-2 is referred to as SARS-CoV -2-S and may for example comprise the amino acid sequence of SEQ ID NO: 403 (1273 amino acids).
- the present disclosure provides exemplary antibodies and antigen-binding antibody fragments that specifically bind to CoV, wherein at least some of these antibodies and antigen-binding antibody fragments specifically bind to SARS-CoV -2-S and/or SARS-CoV-2-S. Due to the sequence similarity among different CoV species, such antibodies or antigen-binding antibody fragments of the present disclosure may also cross react with the S protein of other CoV species.
- the exemplary S proteins of CoV that the antibodies or antigen-binding antibody fragments of the present disclosure may specifically bind include by way of example, Bat SARS CoV (GenBank Accession No. FJ211859), SARS CoV (GenBank Accession No. FJ211860), BtSARS.HKU3.1 (GenBank Accession No. DQ022305), BtSARS.HKU3.2 (GenBank Accession No. DQ084199), BtSARS.HKU3.3 (GenBank Accession No. DQ084200), BtSARS.Rml (GenBank Accession No. DQ412043), BtCoV.279.2005 (GenBank Accession No. DQ648857), BtSARS.Rfl (GenBank Accession No.
- SARSCoV.civetOlO GenBank Accession No. AY572035)
- SARSCoV.MA.15 GenBank Accession No. DQ497008
- Rs SHC014 GenBank® Accession No. KC881005
- Rs3367 GenBank® Accession No. KC881006
- WiVl S GenBank® Accession No. KC881007
- the antibodies and antigen-binding antibody fragments provided herein may also bind to and neutralize existing bat CoV or pre-emergent bat CoVs. Antibodies and antigen binding antibody fragments with such binding and/or neutralization abilities would be particularly useful in a future pandemic that may be caused by a spillover from an animal reservoir, like a bat.
- ADI-55688, ADI-55689, ADI-55993, ADI-5600, ADI-56046, ADI-55690, ADI-56010, and ADI-55951 were shown to neutralize authentic bat coronavirus, WIV1 (see Figure 3E of Wee A. et al, Science. 2020 Jun 15;eabc7424. doi: 10.1126/science.abc7424).
- the S proteins of CoV to which the antibodies or antigen-binding antibody fragments of the present disclosure may specifically bind to and neutralize pre-emergent coronaviruses from other species, e.g., bats.
- the S proteins of CoV to which the antibodies or antigen-binding antibody fragments of the present disclosure may specifically bind to may include, for example, Middle East respiratory syndrome coronavirus isolate Riyadh_2_2012 (GenBank Accession No. KF600652.1), Middle East respiratory syndrome coronavirus isolate Al-Hasa_18_2013 (GenBank Accession No. KF600651.1), Middle East respiratory syndrome coronavirus isolate Al-Hasa_17_2013 (GenBank Accession No. KF600647.1), Middle East respiratory syndrome coronavirus isolate Al-Hasa_15_2013 (GenBank Accession No.
- KF600627.1 Middle East respiratory syndrome coronavirus isolate Bisha_l_2012 (GenBank Accession No. KF600620.1), Middle East respiratory syndrome coronavirus isolate Riyadh_3_2013 (GenBank Accession No. KF600613.1), Middle East respiratory syndrome coronavirus isolate Riyadh_l_2012 (GenBank Accession No. KF600612.1), Middle East respiratory syndrome coronavirus isolate Al-Hasa_3_2013 (GenBank Accession No. KF186565.1), Middle East respiratory syndrome coronavirus isolate Al-Hasa_l_2013 (GenBank Accession No. KF186567.1), Middle East respiratory syndrome coronavirus isolate Al-Hasa_2_2013 (GenBank Accession No.
- KF186566.1 Middle East respiratory syndrome coronavirus isolate Al-Hasa_4_2013 (GenBank Accession No. KF186564.1), Middle East respiratory syndrome coronavirus (GenBank Accession No. KF192507.1), Betacoronavirus England 1-Nl (GenBank Accession No. NC_019843), MERS- CoV_SA-Nl (GenBank Accession No.
- BtCoV.HKU4.1 GenBank Accession No. NC_009019
- BtCoV.HKU4.3 GenBank Accession No. EF065507
- BtCoV.HKU4.4 GenBank Accession No. EF065508
- BtCoV 133.2005 GenBank Accession No. NC 008315
- BtCoV.HKU5.5 GenBank Accession No. EF065512
- BtCoV.HKU5.1 GenBank Accession No. NC_009020
- BtCoV.HKU5.2 GenBank Accession No. EF065510
- BtCoV.HKU5.3 GenBank Accession No.
- the S proteins of CoV to which the antibodies or antigen-binding antibody fragments of the present disclosure may specifically bind may include for example, FCov.FIPV.79.1146.VR.2202 (GenBank Accession No. NV_007025), transmissible gastroenteritis virus (TGEV) (GenBank Accession No. NC_002306; GenBank Accession No. Q811789.2; GenBank Accession No. DQ811786.2; GenBank Accession No. DQ811788.1; GenBank Accession No. DQ811785.1; GenBank Accession No. X52157.1; GenBank Accession No. AJ011482.1; GenBank Accession No. KC962433.1; GenBank Accession No.
- FCov.FIPV.79.1146.VR.2202 GenBank Accession No. NV_007025
- TGEV transmissible gastroenteritis virus
- GenBank Accession No. JQ693060.1 GenBank Accession No. KC609371.1; GenBank Accession No. JQ693060.1; GenBank Accession No. JQ693059.1; GenBank Accession No. JQ693058.1; GenBank Accession No. JQ693057.1; GenBank Accession No. JQ693052.1; GenBank Accession No. JQ693051.1; GenBank Accession No. JQ693050.1), or porcine reproductive and respiratory syndrome virus (PRRSV) (GenBank Accession No. NC_001961.1; GenBank Accession No. DQ811787).
- PRRSV porcine reproductive and respiratory syndrome virus
- the S proteins of CoV to which the antibodies or antigen-binding antibody fragments of the present disclosure may specifically bind may include, for example,
- BtCoV.1 A.AFCD62 GenBank Accession No. NC_010437
- BtCoV.lB.AFCD307 GenBank Accession No. NC_010436
- BtCov.FlKU8.AFCD77 GenBank Accession No. NC_010438
- BtCoV.512.2005 GenBank Accession No. DQ648858
- porcine epidemic diarrhea virus PEDV.CV777 GenBank Accession No. NC_003436, GenBank Accession No. DQ355224.1, GenBank Accession No. DQ355223.1, GenBank Accession No. DQ355221.1, GenBank Accession No. JN601062.1, GenBank Accession No. N601061.1, GenBank Accession No.
- JN601060.1 GenBank Accession No. JN601059.1, GenBank Accession No. JN601058.1, GenBank Accession No. JN601057.1, GenBank Accession No. JN601056.1, GenBank Accession No. JN601055.1, GenBank Accession No. JN601054.1, GenBank Accession No. JN601053.1, GenBank Accession No. JN601052.1, GenBank Accession No. JN400902.1, GenBank Accession No. JN547395.1, GenBank Accession No. FJ687473.1, GenBank Accession No. FJ687472.1, GenBank Accession No. FJ687471.1, GenBank Accession No. FJ687470.1, GenBank Accession No.
- FJ687469.1 GenBank Accession No. FJ687468.1, GenBank Accession No. FJ687467.1, GenBank Accession No. FJ687466.1, GenBank Accession No. FJ687465.1, GenBank Accession No. FJ687464.1, GenBank Accession No. FJ687463.1, GenBank Accession No. FJ687462.1, GenBank Accession No. FJ687461.1, GenBank Accession No. FJ687460.1, GenBank Accession No. FJ687459.1, GenBank Accession No. FJ687458.1, GenBank Accession No. FJ687457.1, GenBank Accession No. FJ687456.1, GenBank Accession No.
- GenBank Accession No. FJ687455.1 GenBank Accession No. FJ687454.1, GenBank Accession No. FJ687453 GenBank Accession No. FJ687452.1, GenBank Accession No. FJ687451.1, GenBank Accession No. FJ687450.1, GenBank Accession No. FJ687449.1, GenBank Accession No. AF500215.1, GenBank Accession No. KF476061.1, GenBank Accession No. KF476060.1, GenBank Accession No. KF476059.1, GenBank Accession No. KF476058.1, GenBank Accession No. KF476057.1, GenBank Accession No. KF476056.1, GenBank Accession No. KF476055.1, GenBank Accession No.
- the S proteins of CoV to which the antibodies or antigen-binding antibody fragments of the present disclosure may specifically bind may include, for example, FtCoV.FlKUl.C.N5 (GenBank Accession No. DQ339101), MFFV.A59 (GenBank Accession No. NC 001846), PHEV.VW572 (GenBank Accession No. NC 007732), HCoV.OC43.ATCC.VR.759 (GenBank Accession No. NC_005147), or bovine enteric coronavirus (BCoV.ENT) (GenBank Accession No. NC_003045).
- the S proteins of CoV to which the antibodies or antigen-binding antibody fragments of the present disclosure may specifically bind may include, for example, BtCoV.HKU9.2 (GenBank Accession No. EF065514), BtCoV.HKU9.1 (GenBank Accession No. NC_009021), BtCoV.HkU9.3 (GenBank Accession No. EF065515), or BtCoV.HKU9.4 (GenBank Accession No. EF065516).
- an anti-CoV-S antibody or antigen-binding fragment thereof binds to CoV-S (e.g., SARS-CoV-S and/or SARS-CoV-2-S, and/or any of the CoV S proteins listed above) with a dissociation constant (KD) of (i) 100 nM or lower; (ii) about 10 nM or lower; (iii) about 1 nM or lower; (iv) about 100 pM or lower; (v) about 10 pM or lower; (vi) about 1 pM or lower; or (vii) about 0.1 pM or lower.
- KD dissociation constant
- the present disclosure provides exemplary antibodies or antigen-binding fragments thereof that bind CoV-S, including human CoV-S, which optionally may be affinity-matured.
- Other antibodies or antigen-binding fragments thereof that bind CoV-S, including those having different CDRs, and epitopic specificity may be obtained using the disclosure of the present specification, and using methods that are generally known in the art.
- Such antibodies and antigen-binding fragments thereof antagonize the biological effects of CoV-S in vivo and therefore are useful in treating or preventing COV-S-related conditions including, particularly coronavirus infection.
- the antibody or antigen-binding fragment thereof according to the disclosure comprises one or more CDRs, a V L chain and/or V H chain of the anti-CoV-S antibodies and antigen-binding fragments thereof described herein.
- an anti-CoV-S antibody or antigen-binding fragment thereof according to the disclosure will interfere with, block, reduce, or modulate the interaction between COV-S and its receptor(s) (e.g., ACE2, CD209L, L-SIGN, DPP4, or CD26) on host cells or a S protein-priming protein on host cells (e.g., TMPRSS2). If binding of the S protein to its receptor is blocked or reduced, CoV virions may be prohibited from entering the cells, i.e., infection to further cells is prevented.
- COV-S and its receptor(s) e.g., ACE2, CD209L, L-SIGN, DPP4, or CD26
- S protein-priming protein on host cells e.g., TMPRSS2
- the S protein is prevented from binding to a S protein-priming protein, the S protein would not be activated and therefore the host cell entry via the receptor may be reduced, i.e., infection to further cells is prevented.
- an anti-CoV-S antibody or antigen-binding fragment thereof according to the disclosure is “neutralizing”, e.g., it substantially or totally prevents the specific interaction of CoV-S with the host receptors or priming protein.
- CoV virions may be substantially or totally cleared by immune cells of the host, such as phagocytes via, for example, Fc receptor mediated phagocytosis or mere phagocytosis due to increased time of virions outside the cells.
- the antibody or antigen-binding fragment thereof neutralizes CoV-S, e.g., by remaining bound to CoV-S in a location and/or manner that prevents CoV-S from specifically binding to its receptor or priming protein on host cells.
- CoV virions may be substantially or totally prevented from entering the cells, i.e. infection to further cells is prevented.
- an anti-CoV-S antibody or antigen-binding fragment thereof neutralizes CoV (e.g., SARS-CoV and/or SARS-CoV-2) at an IC50 of about 100 nM or lower, of about 50 nM or lower, of about 20 nM or lower, of about 10 nM or lower, of about 5 nM or lower, of about 2 nM or lower, of about 1 nM or lower, of about 500 pM or lower, of about 200 pM or lower, of about 100 pM or lower, of about 50 pM or lower, of about 20 pM or lower, of about 10 pM or lower, of about 5 pM or lower, of about 2 pM or lower, or of about 1 pM or lower, or at an IC50 of about 5 ⁇ g/mL or lower, of about 4.5 ⁇ g/mL, of about 4 ⁇ g/mL, of about 3.5 ⁇ g/mL, of about 3
- CoV e.g.
- the isolated antibody or antigen-binding antibody fragment may neutralize SARS-CoV and/or SARS- CoV-2 at an IC90 of about 10 ⁇ g/mL or less, an IC90 of about 1 to about 10 ⁇ g/mL, an IC90 of about 2 to about 10 mg/mL, an IC90 of about 3 to about 10 mg/mL, an IC90 of about 5 to 10 mg/mL, an IC90 of about 7 to 10 mg/mL, an IC90 of about 8 to 10 mg/mL, an IC90 of about 9 to 10 mg/mL, an IC90 of about 10 mg/mL, 9 mg/mL, 8 mg/mL, 7, mg/mL, 6 mg/mL, or 5 mg/mL.
- an anti-CoV-S antibody or antigen-binding fragment thereof according to the disclosure or cocktail thereof when administered to a coronavirus infected host or one susceptible to coronavirus infection such as a health care worker may promote a neutralization response in the host against the coronavirus which is sufficient to permit the host to be able to mount an effective cell-mediated immune response against the virus, e.g., T cell-mediated or cytokine-mediated immune response against the coronavirus and/or to be more responsive to other treatment methods such as drugs, antivirals or other biologies.
- an effective cell-mediated immune response against the virus e.g., T cell-mediated or cytokine-mediated immune response against the coronavirus and/or to be more responsive to other treatment methods such as drugs, antivirals or other biologies.
- the anti-CoV-S antibodies or antigen-binding fragments thereof have a variety of uses.
- the subject antibodies and fragments can be useful in prophylactic or therapeutic applications, as well as diagnostically in binding assays.
- the subject anti-CoV-S antibodies or antigen-binding fragments thereof are useful for affinity purification of CoV-S, in particular human CoV-S or its ligands and in screening assays to identify other antagonists of CoV-S activity.
- Some of the antibodies or antigen-binding fragments thereof are useful for inhibiting binding of CoV-S to its receptor(s) (e.g., ACE2, CD209L, L-SIGN, DPP4, or CD26) on host cells or a S protein-priming protein on host cells (e.g., TMPRSS2) or inhibiting COV-S-mediated activities and/or biological effects.
- its receptor(s) e.g., ACE2, CD209L, L-SIGN, DPP4, or CD26
- S protein-priming protein on host cells e.g., TMPRSS2
- one or more biological effects associated with COV-S refers to any biological effect mediated, induced, or otherwise attributable to COV-S, e.g., binding properties, functional properties, and other properties of biological significance.
- Non-limiting exemplary biological effects of COV-S include COV-S binding to its receptor(s) (e.g., ACE2, CD209L, L-SIGN, DPP4, or CD26) on host cells or a S protein-priming protein on host cells (e.g., TMPRSS2), activation of host cells for allowing virus entry, activation of immune cells as a result of the entry of CoV into the cell, e.g., via presentation of CoV antigen(s) on the host cells’ MHC molecule, and resulting inflammation.
- the subject anti-CoV-S antibodies are capable of inhibiting one, a combination of, or all of these exemplary CoV-S biological activities.
- the anti-CoV-S antibodies and antigen-binding fragments thereof provided herein may neutralize CoV virions or reduce the infectivity of CoV virions.
- the antibody or antigen-binding fragment thereof can be used in a variety of therapeutic applications.
- the anti-CoV-S antibody or antigen-binding fragment thereof are useful for treating conditions associated with CoV-S, such as, but not limited to, symptoms associated with CoV infection.
- the CoV may be any CoV, including SARS-CoV, SARS-CoV-2, MERS-CoV, HCoV-HKUl, HCoV-OC43, HCoV-229E, and HCoV- NL63, and also may be any of the CoV species listed above herein.
- CoV infection-associated symptoms are fever, cough, dry cough, shortness of breath or difficulty of breath, fatigue, aches, runny nose, congestion, sore throat, conjunctivitis, chest pain, headache, muscle ache, chills, loss of smell, and loss of taste, and gastrointestinal symptoms including diarrhea.
- Complications and/or diseases/disorders associated with coronavirus infection may include, for example, bronchitis, pneumonia, respiratory failure, acute respiratory failure, organ failure, multi-organ system failure, pediatric inflammatory multisystem syndrome, acute respiratory distress syndrome (a severe lung condition that causes low oxygen in the blood and organs), blood clots, cardiac conditions, myocardial injury, myocarditis, heart failure, cardiac arrest, acute myocardial infarction, dysrhythmias, venous thromboembolism, post-intensive care syndrome, shock, anaphylactic shock, cytokine release syndrome, septic shock, disseminated intravascular coagulation, ischemic stroke, intracerebral hemorrhage, microangiopathic thrombosis, psychosis, seizure, nonconvulsive status epilepticus, traumatic brain injury, stroke, anoxic brain injury, encephalitis, posterior reversible leukoencephalopathy, necrotizing encephalopathy, post- infectious encepha
- Certain diseases and conditions such as high blood pressure, type 1 diabetes, liver disease, overweight, chronic lung diseases including cystic fibrosis, pulmonary fibrosis, and asthma, compromised immune system due to transplant, use of an immunosuppressant, or HIV infection, and brain and nervous sustem condition, may increase the risk of CoV infection-associated complications and diseases.
- the subject anti-CoV-S antibodies and antigen-binding fragments thereof may be used alone or in association with other active agents or drugs, including other biologies, to treat any subject in which blocking, inhibiting, or neutralizing the in vivo effect of CoV-S or blocking or inhibiting the interaction of CoV-S and its receptor(s) (e.g., ACE2, CD209L, L-SIGN, DPP4, or CD26) on host cells or a S protein-priming protein on host cells (e.g., TMPRSS2), is therapeutically desirable.
- CoV-S and its receptor(s) e.g., ACE2, CD209L, L-SIGN, DPP4, or CD26
- S protein-priming protein on host cells e.g., TMPRSS2
- the subject anti-CoV-S antibody and antigen-binding fragment thereof may be used in combination with a second antibody, or antigen-binding fragment thereof, wherein the second antibody, or antigen-binding fragment thereof, is selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, ADI-58131, or a combination thereof.
- the second antibody, or antigen-binding fragment thereof is ADI-58122.
- the second antibody, or antigen-binding fragment thereof is ADI-58127. In one embodiment, the second antibody, or antigen-binding fragment thereof, is ADI-58129. In one embodiment, the second antibody, or antigen-binding fragment thereof, is ADI-58131. [0396] Exemplary anti-CoV antibodies and antigen-binding fragments thereof according to the disclosure, and the specific CDRs thereof are identified in this section.
- the anti-CoV-S antibodies and antigen-binding fragments thereof comprising the disclosure have binding affinity for CoV-S, such as SARS-CoV-S or SARS-CoV-S2.
- Some antibodies of the present disclosure bind to SARS-CoV-S or SARS-CoV-S2 with a similar K D (M), while some antibodies of the present disclosure bind to SARS-CoV-S with a lower K D (M) ( / ' . e. , higher affinity) than to SARS-CoV-S2, and some antibodies of the present disclosure bind to SARS-CoV-S-2 with a lower K D (M) (i.e., higher affinity) than to SARS-CoV-S.
- Anti-CoV-S antibodies, and antigen-binding fragments thereof, specifically provided herein include: antibodies ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI- 58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, ADI-58131, and antigen-binding fragments thereof.
- Any Fc variant including but not limited to those specifically disclosed in Table 9 may be used in combination with any of the variable sequences disclosed herein.
- the Fc variant is an FA variant and comprises the amino acid sequence of SEQ ID NO: 413.
- the antibody ADI-58125 comprises an Fc variant of SEQ ID NO:413.
- Tables 1-8 show the SEQ ID NOs assigned to individual amino acid sequences of the HC, VH, VH FR1, VH CDR1, VH FR2, VH CDR2, VH FR3, VH CDR3, VH FR4, EC, VL, VL FR1, VL CDR1, VL FR2, VL CDR2, VL FR3, VL CDR3, and VL FR4 for individual antibodies, and the SEQ ID NOs assigned to the nucleic acid sequences of the VH and VL of individual antibodies.
- anti-CoV-S antibodies or antigen-binding antibody fragments comprising (i) a VH CDR that is same as the VH CDR3 of, (ii) a VH CDR3 and VL CDR3, both of which as same as both of the VH CDR3 and the VL CDR3 of, (iii) at least 1, 2, 3, 4, 5, or 6 CDRs that are same as the corresponding CDR(s) of, or (iv) 6 CDRs that are all the same as the 6 CDRs of any one of the disclosed antibodies selected from the group consisting of ADI-58120, ADI- 58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI- 58129, ADI-58130, and ADI-58
- anti-CoV-S antibodies or antigen-binding antibody fragments which optionally may be affinity-matured, comprising one of the CDR requirements (i)-(iv) of the immediately above paragraph, further wherein (a) the VH comprises an amino acid sequence with at least 80, 85, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of the VH of, and (b) the VL comprises an amino acid sequence with at least 80, 85, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of the VL of any one of the disclosed antibodies selected from the group consisting of ADI- 58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI- 58128,
- the disclosure contemplates anti-CoV-S antibodies or antigen binding antibody fragments which optionally may be affinity-matured, comprising one of the VH and VL requirements (i)-(iv) of the immediately above paragraph, further wherein (a) the heavy chain comprises an amino acid sequence with at least 80, 85, 90, 91, 92, 93, 9495, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of the heavy chain of, and (b) the light chain comprises an amino acid sequence with at least 80, 85, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of the light chain of any one of the disclosed antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI- 58125, ADI-58126, ADI-58127, ADI-58128,
- the disclosure contemplates anti-CoV-S antibodies or antigen binding antibody fragments which optionally may be affinity-matured, comprising one of the CDR requirements (i)-(iv) of the immediately above paragraph, further wherein (a) the VH is identical to the VH of, and (b) the VL is identical to the VL of any one of the disclosed antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, and ADI-58131.
- the disclosure includes antibodies and antigen-binding fragments which optionally may be affinity-matured, having binding specificity to COV-S that bind the same epitope as one of antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI- 58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI- 58130, and ADI-58131.
- the disclosure includes antibodies and antigen-binding fragments having binding specificity to COV-S, which optionally may be affinity-matured, that bind the same epitope as any one of antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI- 58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI- 58130, and ADI-58131.
- the anti-CoV-S antibodies and antigen-binding fragments optionally may be affinity-matured, comprise, or alternatively consist of, combinations of one or more of the FRs, CDRs, the VH and VL sequences, and the heavy chain and light chain sequences set forth above, including all of them, or sequences that are at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical thereto.
- antigen-binding fragments comprise, or alternatively consist of, Fab fragments having binding specificity for CoV-S.
- the Fab fragment preferably includes the VH and the VL sequence of any one of antibodies selected from the group consisting of ADI-58120, ADI- 58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI- 58129, ADI-58130, and ADI-58131, or sequences that are at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical thereto.
- This embodiment further includes Fabs containing additions, deletions, and variants of such VH and VL sequence while retaining binding specificity for CoV-S.
- Fab fragments may be produced by enzymatic digestion (e.g., papain) of the parent full antibody.
- anti-CoV-S antibodies such as anyone of antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, and ADI- 58131, and Fab fragments thereof may be produced via expression in mammalian cells, such as CHO, NSO, or HEK 293 cells, fungal, insect, or microbial systems, such as yeast cells.
- polynucleotides encoding antibody polypeptides having binding specificity to COV-S including the VH and VL of any one of antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, and ADI-58131, as well as fragments, variants, optionally affinity-matured variants, and combinations of one or more of the FRs, CDRs, the VH and VL sequences, and the heavy chain and light chain sequences set forth above, including all of them, or sequences that are at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%,
- the disclosure contemplates isolated anti-CoV-S antibodies and antigen binding fragments comprising (i) a VH which is same as the VH of any one of antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, and ADI-58131; and (ii) a VL which is same as the VL of another antibody selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, and ADI-58131, or a variant thereof, wherein optionally one or more of the framework region residues (“LR residues”) and/or
- the disclosure also includes humanized, primatized and other chimeric forms of these antibodies.
- the chimeric and humanized antibodies may include an Lc derived from IgGl, IgG2,
- IgG3, or IgG4 constant regions are constant regions.
- the chimeric or humanized antibodies or fragments or VH or VL polypeptides originate or are derived from one or more human antibodies, e.g., a human antibody identified from a clonal human B cell population.
- the disclosure provides vectors comprising a nucleic acid molecule encoding an anti-CoV-S antibody or fragment thereof as disclosed herein.
- the disclosure provides host cells comprising a nucleic acid molecule encoding an anti-CoV-S antibody or fragment thereof as disclosed herein.
- the disclosure provides isolated antibodies or antigen binding fragments thereof that competes for binding to CoV-S with an antibody or antigen binding fragment thereof disclosed herein.
- the disclosure provides a nucleic acid molecule encoding any of the antibodies or antigen binding fragments disclosed herein.
- the disclosure provides a pharmaceutical or diagnostic composition comprising at least one antibody or antigen binding fragment thereof as disclosed herein.
- the disclosure provides a method for treating or preventing a condition associated with elevated CoV-S levels in a subject, comprising administering to a subject in need thereof an effective amount of at least one isolated antibody or antigen binding fragment thereof as disclosed herein.
- the disclosure provides a method of inhibiting binding of COV-S to its receptor (e.g., ACE2, L-SIGN, CD209L, DPP4, CD26) or an S protein-priming protein (e.g., TMPRSS2) in a subject comprising administering an effective amount of at least one antibody or antigen binding fragment thereof as disclosed herein.
- its receptor e.g., ACE2, L-SIGN, CD209L, DPP4, CD26
- S protein-priming protein e.g., TMPRSS2
- administering one or more of antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, and ADI- 58131 may inhibit binding of COV-S to its receptor, e.g., ACE2.
- the disclosure provides an antibody or antigen binding fragment thereof that selectively binds to CoV-S, wherein the antibody or antigen binding fragment thereof binds to CoV-S with a K D of less than or equal to 5x10 5 M, 10 5 M, 5x10 6 M, 10 6 M, 5x10 7 M, 10 7 M, 5x10 8 M,
- the anti-CoV-S antibody or antigen binding fragment has cross-reactivity to the S protein of CoV other than SARS-CoV-S or SARS-CoV-2-S.
- inventive antibodies and antigen binding fragments thereof may be modified post- translationally to add effector moieties such as chemical linkers, detectable moieties such as for example fluorescent dyes, enzymes, substrates, bioluminescent materials, radioactive materials, and chemiluminescent moieties, or functional moieties such as for example streptavidin, avidin, biotin, a cytotoxin, a cytotoxic agent, and radioactive materials.
- effector moieties such as chemical linkers, detectable moieties such as for example fluorescent dyes, enzymes, substrates, bioluminescent materials, radioactive materials, and chemiluminescent moieties, or functional moieties such as for example streptavidin, avidin, biotin, a cytotoxin, a cytotoxic agent, and radioactive materials.
- Antibodies and antigen binding fragments thereof may also be chemically modified to provide additional advantages such as increased solubility, stability and circulating time (in vivo half- life) of the polypeptide, or decreased immunogenicity ( See U.S. Patent No. 4,179,337).
- the chemical moieties for derivatization may be selected from water soluble polymers such as polyethylene glycol, ethylene glycol/propylene glycol copolymers, carboxymethylcellulose, dextran, polyvinyl alcohol, and the like.
- the antibodies and fragments thereof may be modified at random positions within the molecule, or at predetermined positions within the molecule and may include one, two, three, or more attached chemical moieties.
- the polymer may be of any molecular weight, and may be branched or unbranched.
- the preferred molecular weight is between about 1 kDa and about 100 kDa (the term "about” indicating that in preparations of polyethylene glycol, some molecules will weigh more, some less, than the stated molecular weight) for ease in handling and manufacturing.
- Other sizes may be used, depending on the desired therapeutic profile (e.g., the duration of sustained release desired, the effects, if any on biological activity, the ease in handling, the degree or lack of antigenicity and other known effects of the polyethylene glycol to a therapeutic protein or analog).
- the polyethylene glycol may have an average molecular weight of about 200, 500, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 5500, 6000, 6500, 7000, 7500, 8000, 8500, 9000, 9500, 10,000,
- Branched polyethylene glycols are described, for example, in U.S. Patent No. 5,643,575; Morpurgo et al., Appl. Biochem. Biotechnol., 56:59-72 (1996); Vorobjev et al, Nucleosides and Nucleotides, 18:2745-2750 (1999); and Caliceti et al, Bioconjug. Chem., 10:638-646 (1999), the disclosures of each of which are incorporated herein by reference.
- polyethylene glycol may be covalently bound through amino acid residues via a reactive group, such as, a free amino or carboxyl group.
- Reactive groups are those to which an activated polyethylene glycol molecule may be bound.
- the amino acid residues having a free amino group may include lysine residues and the N-terminal amino acid residues; those having a free carboxyl group may include aspartic acid residues glutamic acid residues and the C-terminal amino acid residue.
- Sulfhydryl groups may also be used as a reactive group for attaching the polyethylene glycol molecules. Preferred for therapeutic purposes is attachment at an amino group, such as attachment at the N-terminus or lysine group.
- polyethylene glycol may be attached to proteins via linkage to any of a number of amino acid residues.
- polyethylene glycol can be linked to polypeptides via covalent bonds to lysine, histidine, aspartic acid, glutamic acid, or cysteine residues.
- One or more reaction chemistries may be employed to attach polyethylene glycol to specific amino acid residues (e.g., lysine, histidine, aspartic acid, glutamic acid, or cysteine) or to more than one type of amino acid residue (e.g., lysine, histidine, aspartic acid, glutamic acid, cysteine and combinations thereof).
- antibodies or antigen binding fragments thereof having increased in vivo half- lives may be produced via fusion with albumin (including but not limited to recombinant human serum albumin or fragments or variants thereof (See, e.g., U.S. Patent No. 5,876,969, EP 0413 622, and U.S. Patent No. 5,766,883, herein incorporated by reference in their entirety)), or other circulating blood proteins such as transferrin or ferritin.
- polypeptides and/or antibodies of the present disclosure are fused with the mature form of human serum albumin (i.e., amino acids 1-585 of human serum albumin as shown in FIGS.
- detectable moieties include, but are not limited to, horseradish peroxidase, acetylcholinesterase, alkaline phosphatase, Z3 ⁇ 4Za-galactosidase, and luciferase.
- detectable moieties include, but are not limited to, horseradish peroxidase, acetylcholinesterase, alkaline phosphatase, Z3 ⁇ 4Za-galactosidase, and luciferase.
- Further exemplary fluorescent materials include, but are not limited to, rhodamine, fluorescein, fluorescein isothiocyanate, umbelliferone, dichlorotriazinylamine, phycoerythrin, and dansyl chloride.
- chemiluminescent moieties include, but are not limited to, luminol.
- Further exemplary bioluminescent materials include, but are not limited to, luciferin and aequorin.
- Further exemplary radioactive materials include, but are not limited to, Iodine 125 ( 125 I), Carbon 14 ( 14 C), Sulfur 35 ( 35 S), Tritium ( 3 H) and Phosphorus 32 ( 32 P).
- Embodiments described herein further include variants and equivalents that are substantially homologous to the antibodies, antibody fragments, diabodies, SMIPs, camelbodies, nanobodies, IgNAR, polypeptides, variable regions, and CDRs set forth herein.
- These may contain, e.g., conservative substitution mutations, (i.e., the substitution of one or more amino acids by similar amino acids).
- conservative substitution refers to the substitution of an amino acid with another within the same general class, e.g., one acidic amino acid with another acidic amino acid, one basic amino acid with another basic amino acid, or one neutral amino acid by another neutral amino acid.
- conservative amino acid substitution is well known in the art.
- the disclosure contemplates polypeptide sequences having at least 90% or greater sequence homology to any one or more of the polypeptide sequences of antigen binding fragments, variable regions and CDRs set forth herein. More preferably, the disclosure contemplates polypeptide sequences having at least 95% or greater sequence homology, even more preferably at least 98% or greater sequence homology, and still more preferably at least 99% or greater sequence homology to any one or more of the polypeptide sequences of antigen binding fragments, variable regions, and CDRs set forth herein.
- the disclosure further contemplates the above-recited polypeptide homologs of the antigen binding fragments, variable regions and CDRs set forth herein further having anti-CoV-S activity.
- anti-CoV-S activity are set forth herein, e.g., ability to inhibit CoV-S binding to its receptor such as ACE2 or L-SIGN or an S protein-priming protein, thereby resulting in the reduced entry of CoV into cells.
- the disclosure further contemplates the generation and use of antibodies that bind any of the foregoing sequences, including, but not limited to, anti-idiotypic antibodies.
- an anti-idiotypic antibody could be administered to a subject who has received an anti-CoV-S antibody to modulate, reduce, or neutralize, the effect of the anti-CoV-S antibody.
- Such antibodies could also be useful for treatment of an autoimmune disease characterized by the presence of anti-CoV-S antibodies.
- a further exemplary use of such antibodies, e.g., anti-idiotypic antibodies, is for detection of the anti-CoV-S antibodies of the present disclosure, for example to monitor the levels of the anti-CoV-S antibodies present in a subject’s blood or other bodily fluids.
- the disclosure provides a method of using the anti- idiotypic antibody to monitor the in vivo levels of said anti-CoV-S antibody or antigen binding fragment thereof in a subject or to neutralize said anti-CoV-S antibody in a subject being administered said anti-CoV-S antibody or antigen binding fragment thereof.
- the present disclosure also contemplates anti-CoV-S antibodies comprising any of the polypeptide or polynucleotide sequences described herein substituted for any of the other polynucleotide sequences described herein.
- the present disclosure contemplates antibodies comprising the combination of any of the VF and VH sequences described herein, and further contemplates antibodies resulting from substitution of any of the CDR sequences described herein for any of the other CDR sequences described herein.
- Another embodiment of the disclosure contemplates these polynucleotides incorporated into an expression vector for expression in mammalian cells such as CHO, NSO, or HEK-293 cells, or in fungal, insect, or microbial systems such as yeast cells.
- Fab fragments can be produced by enzymatic digestion (e.g., papain) of any one of antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, and ADI-58131, following expression of the full-length polynucleotides in a suitable host.
- enzymatic digestion e.g., papain
- anti-Co V-S antibodies such as anyone of antibodies selected from the group consisting of ADI- 58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI- 58128, ADI-58129, ADI-58130, and ADI-58131, or Fab fragments thereof, can be produced via expression of the polynucleotides encoding any one of antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI- 58127, ADI-58128, ADI-58129, ADI-58130, and ADI-58131, in mammalian cells such as CFIO,
- NSO or F1EK 293 cells
- fungal or insect
- microbial systems such as yeast cells.
- Flost cells and vectors comprising said polynucleotides are also contemplated.
- the disclosure further contemplates vectors comprising the polynucleotide sequences encoding the variable heavy and light chain polypeptide sequences, as well as the individual CDRs (hypervariable regions), as set forth herein, as well as host cells comprising said vector sequences.
- the host cells are mammalian cells, such as CFIO cells.
- the host cells are yeast cells.
- the disclosure is further directed to antibody-drug conjugates (ADCs) comprising (a) any antibody or antigen-binding antibody fragment described herein; and (b) a drug conjugated to the antibody or antigen-binding antibody fragment, either directly or indirectly (e.g., via a linker).
- ADCs antibody-drug conjugates
- the drug may be, but not limited to, a cytotoxic drug, an apoptotic drug, an immunostimulatory drug, an anti-microbial drug, an antibacterial drug or vaccine, an antiviral drug, antihelminth drug, antiparasitic drug, an anti-inflammatory drug, antihistamine, an anti-fibrotic drug, an immunosuppressive drug, a steroid, a bronchodilator, a beta blocker, an ACE inhibitor, an enzyme, a serine protease inhibitor, a toxin, a radioisotope, a compound, a small molecule, a small molecule inhibitor, a protein, a peptide, a vector, a plasmid, a viral particle, a nanoparticle, a DNA molecule, an RNA molecule, an siRNA, an shRNA, a micro RNA, an oligonucleotide, and an imaging drug.
- An antiviral drug may be remdesivir, favipiravir, darunavir, nelfinavir, saquinavir, lopinavir or ritonavir; an antihelminth drug may be ivermectin; an antiparasite drug may be hydroxychloroquine, chloroquine, or atovaquone; antibacterial drug or vaccine may be the tuberculosis vaccine BCG; an anti-inflammatory drug, may be ciclesonide, a TNF inhibitor (e.g., adalimumab), a TNF receptor inhibitor (e.g., etanercept), an IL-6 inhibitor (e.g., clazakizumab), an IL-6 receptor inhibitor (e.g., toclizumab), or metamizole; an antihistamine drug may be bepotastine; an ACE inhibitor may be moexipril; and a drug that inhibits priming of CoV-S may be
- the toxin may be a bacterial, fungal, plant, or animal toxin, or a fragment thereof.
- examples include, but are not limited to, diphtheria A chain, diphtheria toxin, exotoxin A chain, ricin A chain, abrin A chain, modeccin A chain, alpha sarcin, Aleurites fordii protein, a dianthin protein, or a Phytolacca Americana protein.
- the cytotoxic drug or anti-proliferative drug may be, for example, but is not limited to, doxorubicin, daunorubicin, cucurbitacin, chaetocin, chaetoglobosin, chlamydocin, calicheamicin, nemorubicin, cryptophyscin, mensacarcin, ansamitocin, mitomycin C, geldanamycin, mechercharmycin, rebeccamycin, safracin, okilactomycin, oligomycin, actinomycin, sandramycin, hypothemycin, polyketomycin, hydroxyellipticine, thiocolchicine, methotrexate, triptolide, taltobulin, lactacystin, dolastatin, auristatin, monomethyl auristatin E (MMAE), monomethyl auristatin F (MMAF), telomestatin, tubastatin A, combreta
- the radioisotope may be for example, but is not limited to, At 211 , 1 131 , In 131 , 1 125 , Y 90 , Re 186 , Re 188 , Sm 153 , Bi 212 , P 32 , Pb 212 and radioactive isotopes of Lu.
- the drug may be, but is not limited to, MMAE or MMAF.
- the Ab or antigen-binding Ab fragment is directly conjugated to the drug to form an ADC.
- the antibody or antigen-binding antibody fragment is indirectly conjugated to the drug to form an ADC.
- Any appropriate conjugation method may be used to generate an ADC (for example, Nolting B. Methods Mol Biol. 2013;1045:71-100; Jain N. etal., Pharm Res. 2015 Nov;32(ll):3526-40; Tsuchikama K. et al, Protein Cell. 2018 Jan;9(l):33-46; Polakis P. et ai, Pharmacol Rev . 2016 Jan;68(l):3-19).
- methods that may be used to perform conjugation include, but are not limited to, chemical conjugation and enzymatic conjugation.
- Chemical conjugation may utilize, for example, but is not limited to, lysine amide coupling, cysteine coupling, and/or non-natural amino acid incorporation by genetic engineering.
- Enzymatic conjugation may utilize, for example, but is not limited to, transpeptidation using sortase, transpeptidation using microbial transglutaminase, and/or N-Glycan engineering.
- one or more of cleavable linkers may be used for conjugation.
- the cleavable linker may enable cleavage of the drug upon responding to, for example, but not limited to, an environmental difference between the extracellular and intracellular environments (pH, redox potential, etc.) or by specific lysosomal enzymes.
- cleavable linker examples include, but are not limited to, hydrazone linkers, peptide linkers including cathepsin B-responsive linkers, such as valine -citrulline (vc) linker, disulfide linkers such as N-succinimidyl-4-(2-pyridyldithio) (SPP) linker or N-succinimidyl-4-(2- pyridyldithiojbutanoate (SPDB) linker, and pyrophosphate diester linkers.
- peptide linkers including cathepsin B-responsive linkers, such as valine -citrulline (vc) linker
- disulfide linkers such as N-succinimidyl-4-(2-pyridyldithio) (SPP) linker or N-succinimidyl-4-(2- pyridyldithiojbutanoate (SPDB) linker
- SPDB
- non-cleavable linkers may be used.
- non-cleavable linkers include thioether linkers, such as N-succinimidyl 4-(N-maleimidomethyl) cyclohexane- 1-carboxylate (SMCC), and maleimidocaproyl (me) linkers.
- thioether linkers such as N-succinimidyl 4-(N-maleimidomethyl) cyclohexane- 1-carboxylate (SMCC), and maleimidocaproyl (me) linkers.
- SMCC N-succinimidyl 4-(N-maleimidomethyl) cyclohexane- 1-carboxylate
- me maleimidocaproyl
- a compound specific to CoV-S may be a chimeric antigen receptor (CAR).
- the CARs of the present disclosure comprise an antigen binding (AB) domain that binds to CoV-S, a transmembrane (TM) domain, and an intracellular signaling (ICS) domain.
- AB antigen binding
- TM transmembrane
- ICS intracellular signaling
- a CAR may comprise a hinge that joins the AB domain and said TM domain.
- the CAR may comprise one or more costimulatory (CS) domains.
- a CAR according to the disclosure will comprise an antigen-binding (AB) domain which binds to COV-S.
- the AB domain of the CAR may comprise any of the anti- COV-S antigen-binding antibody fragments disclosed herein.
- the AB domain of the CAR may comprise any of the antigen-binding domain of any of the anti-COV-S antibodies disclosed herein.
- the AB domain of the CAR may comprise any of the anti-COV-S antibodies, anti-COV-S antigen-binding antibody fragments, anti-COV-S multi-specific Abs, anti- COV-S multi-specific antigen-binding antibody fragments, and anti-COV-S ADCs disclosed herein, or the ABD thereof.
- the AB domain of the CAR may comprise an anti-COV-S scFv.
- the AB domain may comprise an amino acid sequence at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical to an scFv comprising the VH and VL of any one of antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI- 58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI- 58130, and ADI-58131.
- the AB domain may compete for binding to COV-S with any one of antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, and ADI- 58131.
- the CAR may comprise a hinge sequence between the AB domain and the TM domain.
- a hinge sequence is a short sequence of amino acids that facilitates flexibility (see, e.g. Woof J.M. et al, Nat. Rev. Immunol.,
- the hinge sequence can be any suitable sequence derived or obtained from any suitable molecule.
- the length of the hinge sequence may be optimized based on the desired length of the extracellular portion of the CAR, which may be based on the location of the epitope within the target molecule. For example, if the epitope is in the membrane proximal region within the target molecule, longer hinges may be optimal.
- the hinge may be derived from or include at least a portion of an immunoglobulin Fc region, for example, an IgGl Fc region, an IgG2 Fc region, an IgG3 Fc region, an IgG4 Fc region, an IgE Fc region, an IgM Fc region, or an IgA Fc region.
- the hinge includes at least a portion of an IgGl, an IgG2, an IgG3, an IgG4, an IgE, an IgM, or an IgA immunoglobulin Fc region that falls within its CH2 and CH3 domains.
- the hinge may also include at least a portion of a corresponding immunoglobulin hinge region.
- the hinge is derived from or includes at least a portion of a modified immunoglobulin Fc region, for example, a modified IgGl Fc region, a modified IgG2 Fc region, a modified IgG3 Fc region, a modified IgG4 Fc region, a modified IgE Fc region, a modified IgM Fc region, or a modified IgA Fc region.
- the modified immunoglobulin Fc region may have one or more mutations (e.g., point mutations, insertions, deletions, duplications) resulting in one or more amino acid substitutions, modifications, or deletions that cause impaired binding of the hinge to an Fc receptor (FcR).
- the modified immunoglobulin Fc region may be designed with one or more mutations which result in one or more amino acid substitutions, modifications, or deletions that cause impaired binding of the hinge to one or more FcR including, but not limited to, FcyRI, FcyR2A, FcyR2Bl, Fcy2B2, Fey 3A, Fey 3B, FceRI, FcsR2, FcaRI, Fca/pR, or FcRn.
- a portion of the immunoglobulin constant region may serve as a hinge between the AB domain, for example scFv or nanobody, and the TM domain.
- the hinge can be of a length that provides for increased responsiveness of the CAR-expressing cell following antigen binding, as compared to in the absence of the hinge. In some examples, the hinge is at or about 12 amino acids in length or is no more than 12 amino acids in length.
- Exemplary hinges include those having at least about 10 to 229 amino acids, about 10 to 200 amino acids, about 10 to 175 amino acids, about 10 to 150 amino acids, about 10 to 125 amino acids, about 10 to 100 amino acids, about 10 to 75 amino acids, about 10 to 50 amino acids, about 10 to 40 amino acids, about 10 to 30 amino acids, about 10 to 20 amino acids, or about 10 to 15 amino acids, and including any integer between the endpoints of any of the listed ranges.
- a hinge has about 12 amino acids or less, about 119 amino acids or less, or about 229 amino acids or less.
- Exemplary hinges include a CD28 hinge, IgG4 hinge alone, IgG4 hinge linked to CH2 and CH3 domains, or IgG4 hinge linked to the CH3 domain.
- Exemplary hinges include, but are not limited to, those described in Hudecek M. et al. (2013) Clin. Cancer Res., 19:3153, international patent application publication number WO2014031687, U.S. Pat. No. 8,822,647 or published App. No. US2014/0271635.
- Known hinge sequences include those derived from CD8 a molecule or a CD28 molecule.
- the CAR can be designed to comprise a TM domain that is fused to the AB domain of the CAR.
- a hinge sequence may be inserted between the AB domain and the TM domain.
- TM domains may be derived from a natural or from synthetic sources. Where the source is natural, the domain may be derived from any membrane-bound or transmembrane protein.
- a TM domain denotes a single transmembrane a helix of a transmembrane protein, also known as an integral protein.
- TM domains e.g., may be derived from (i.e.
- the TM domain may be synthetic, in which case the TM domain will comprise predominantly hydrophobic residues such as leucine and valine. Preferably a triplet of phenylalanine, tryptophan and valine will be found at each end of a synthetic TM domain.
- a TM domain is generally thermodynamically stable in a membrane. It may be a single a helix, a transmembrane b barrel, a b- helix of gramicidin A, or any other structure. Transmembrane helices are usually about 20 amino acids in length.
- a well-used TM domain comprises the TM region of CD28, e.g., human CD28. Often, a short oligo- or polypeptide spacer, e.g., between 2 and 10 amino acids in length is used to form the linkage between the TM domain and the ICS domain(s) of the CAR.
- Intracellular signaling ) domain and costimulatorv (CS ) domain [0466]
- the ICS domain or the cytoplasmic domain of a CAR generally triggers or elicits activation of at least one of the normal effector functions of the cell in which the CAR has been placed.
- effector function refers to a specialized function of a cell.
- Effector function of a T cell may be cytolytic activity or helper activity including the secretion of cytokines.
- intracellular signaling domain or “ICS domain” refers to the portion of a protein which transduces the effector function signal and directs the cell to perform a specialized function. While usually the entire ICS domain can be employed, in many cases it is not necessary to use the entire chain. To the extent that a truncated portion of the intracellular signaling domain is used, such truncated portion may be used in place of the intact chain as long as it transduces the effector function signal.
- intracellular signaling domain or “ICS domain” is thus meant to include any truncated portion of the ICS domain sufficient to transduce the effector function signal.
- ICS domains include the cytoplasmic sequences of the T cell receptor (TCR) and co-receptors that act in concert to initiate signal transduction following antigen receptor engagement, as well as any derivative or variant of these sequences and any synthetic sequence that has the same functional capability.
- TCR T cell receptor
- CS domain costimulatory domain
- a CS domain is a domain that transduces such a secondary or costimulatory signal.
- a CAR of the present disclosure may comprise two or more CS domains. The CS domain(s) may be placed upstream of the ICS domain or downstream of the ICS domain.
- T cell activation can be mediated by two distinct classes of cytoplasmic signaling sequence: those that initiate antigen-dependent primary activation through the TCR (primary cytoplasmic signaling sequences) and those that act in an antigen-independent manner to provide a secondary or costimulatory signal (secondary cytoplasmic signaling sequences).
- Primary cytoplasmic signaling sequences regulate primary activation of the TCR complex either in a stimulatory way, or in an inhibitory way.
- Primary cytoplasmic signaling sequences that act in a stimulatory manner may contain signaling motifs which are known as immunoreceptor tyrosine-based activation motifs or IT AMs.
- Such a cytoplasmic signaling sequence may be contained in the ICS or the CS domain of a CAR.
- ITAM-containing primary cytoplasmic signaling sequences include those derived from an ICS domain of a lymphocyte receptor chain, a TCR/CD3 complex protein, an Fc receptor subunit, an IL-2 receptor subunit, CD3z, FcR g, FcR , CD3y, CD35, CD3e, CD5, CD22, CD66d, CD79a, CD79b, CD278 (ICOS), Fee RI, DAP10, and DAP12.
- a well-used ICS domain comprises a cytoplasmic signaling sequence derived from CD3 zeta.
- the CD3z ICS domain may be combined with one or more of other cytoplasmic domain(s).
- the cytoplasmic domain of the CAR can comprise a CD3 z ICS domain and a CS domain wherein a CS region refers to a portion of the CAR comprising the intracellular domain of a costimulatory molecule.
- a costimulatory molecule is a cell surface molecule other than an antigen receptor or their ligands that is required for an efficient response of lymphocytes to an antigen.
- co-stimulatory molecules include an MHC class I molecule, TNF receptor proteins, Immunoglobulin-like proteins, cytokine receptors, integrins, signaling lymphocytic activation molecules (SLAM proteins), activating NK cell receptors, a Toll ligand receptor, B7-H3, BAFFR, BTLA, BLAME (SLAMF8), CD2, CD4, CD5, CD7, CD 8 a, CD 8 b, CDlla, LFA-1 (CDlla/CD18), CDllb, CDllc, CDlld, CD18, CD19, CD19a, CD27, CD28, CD29, CD30, CD40, CD49a, CD49D, CD49f, CD69, CD84, CD96 (Tactile), CD100 (SEMA4D), CD103, CRTAM, 0X40 (CD 134), 4-1BB (CD137), SLAM (SLAMF1, CD150, IPO-3), CD160 (BY55), SELPLG (MHC class I
- a CAR construct may comprise the following format: “AB domain - hinge - TM domain - CS domain - ICS domain.”
- CARs may comprise an amino acid sequence at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical to any of the exemplary constructs below.
- the “anti-CoV-S scFv” may be an scFv generated by linking the VH and VL (in the order of VH-linker-VL or VL-linker-VH) of any one of anti-CoV-S antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, and ADI-58131.
- a leader sequence may be placed upstream of the polynucleotide sequences encoding the CAR.
- the leader sequence facilitates the expression of the CAR on the cell surface.
- CARs according to the present disclosure nucleotide sequences encoding the same, vectors encoding the same, and cells comprising nucleotide sequences encoding said CARs may be further modified, engineered, optimized, or appended in order to provide or select for various features. These features may include, but are not limited to, efficacy, persistence, target specificity, reduced immunogenicity, multi-targeting, enhanced immune response, expansion, growth, reduced off-target effect, reduced subject toxicity, improved target cytotoxicity, improved attraction of disease alleviating immune cells, detection, selection, targeting, and the like.
- the cells may be engineered to express another CAR, or to have a suicide mechanism, and may be modified to remove or modify expression of an endogenous receptor or molecule such as a TCR and/or MHC molecule.
- the vector or nucleic acid sequence encoding the CAR further encodes other genes.
- the vector or nucleic acid sequence may be constructed to allow for the co-expression of multiple genes using a multitude of techniques including co-transfection of two or more plasmids, the use of multiple or bidirectional promoters, or the creation of bicistronic or multicistronic vectors.
- multicistronic vectors may include the encoding of IRES elements or 2 A peptides, such as T2A, P2A, E2A, or F2A (for example, see Kim, J.H., et ai, “High cleavage efficiency of a 2A peptide derived from porcine teschovirus-1 in human cell lines, zebrafish and mice”, PLoS One.
- the CAR expressing cell may further comprise a disruption to one or more endogenous genes.
- the CARs of the present disclosure and cells expressing these CARs may be further modified to improve efficacy against cells expressing the target molecule.
- the cells may be cells expressing COV-S.
- the cells expressing COV-S may be cancer cells, vascular cells, or any other target disease- associated cells.
- the improved efficacy may be measured by increased cytotoxicity against cells expressing the target molecule, for example cytotoxicity against cancer cells.
- the improved efficacy may also be measured by increased production of cytotoxic mediators such as, but not limited to, IFN g, perforin, and granzyme B.
- the improved efficacy may be shown by reduction in the signature cytokines of the diseases, or alleviated symptoms of the disease when the CAR expressing cells are administered to a subject.
- Other cytokines that may be reduced include TGF-beta, IL-6, IL-4, IL-10, and/or IL-13.
- the improved efficacy may be shown by COV-S-specific immune cell responses, such as T cell cytotoxicity.
- improved efficacy may be shown by better tumor cytotoxicity, better infiltration into the tumor, reduction of immunosuppressive mediators, reduction in weight decrease, reduction in ascites, reduction in tumor burden, and/or increased lifespan.
- reduced responsiveness of autoreactive cells or decrease in autoreactive T cells, B cells, or Abs may represent improved efficacy.
- gene expression profiles may be also investigated to evaluate the efficacy of the CAR.
- the CAR expressing cells are further modified to evade or neutralize the activity of immunosuppressive mediators, including, but not limited to prostaglandin E2 (PGE2) and adenosine.
- this evasion or neutralization is direct.
- this evasion or neutralization is mediated via the inhibition of protein kinase A (PKA) with one or more binding partners, for example ezrin.
- PGE2 prostaglandin E2
- the CAR-expressing cells further express the peptide “regulatory subunit I anchoring disruptor” (RIAD).
- RIAD is thought to inhibit the association of protein kinase A (PKA) with ezrin, which thus prevents PKA’s inhibition of TCR activation (Newick K. et al. Cancer Immunol Res. 2016 Jun;4(6):541-51).
- PKA protein kinase A
- the CAR expressing cells may induce a broad immune response, consistent with epitope spreading.
- the CAR expressing cells further comprise a homing mechanism.
- the cell may transgenically express one or more stimulatory chemokines or cytokines or receptors thereof.
- the cells are genetically modified to express one or more stimulatory cytokines.
- one or more homing mechanisms are used to assist the inventive cells to accumulate more effectively to the disease site.
- the CAR expressing cells are further modified to release inducible cytokines upon CAR activation, e.g., to attract or activate innate immune cells to a targeted cell (so-called fourth generation CARs or TRUCKS).
- CARs may co-express homing molecules, e.g., CCR4 or CCR2b, to increase trafficking to the disease site.
- apoptosis using, e.g., a caspase fused to a dimerization domain (see, e.g., Di et al, N Engl. J. Med. 2011 Nov. 3; 365(18): 1673-1683), can be used as a safety switch in the CAR therapy of the instant disclosure.
- CAR-expressing cells can also express an inducible Caspase-9 (iCaspase-9) molecule that, upon administration of a dimerizer drug (e.g., rimiducid (also called AP1903 (Bellicum Pharmaceuticals) or AP20187 (Ariad)) leads to activation of the Caspase-9 and apoptosis of the cells.
- a dimerizer drug e.g., rimiducid (also called AP1903 (Bellicum Pharmaceuticals) or AP20187 (Ariad)
- AP1903 also called AP1903 (Bellicum Pharmaceuticals)
- AP20187 AP20187
- the iCaspase-9 molecule is encoded by a nucleic acid molecule separate from the CAR-encoding vector(s). In some cases, the iCaspase-9 molecule is encoded by the same nucleic acid molecule as the CAR-encoding vector.
- the iCaspase-9 can provide a safety switch to avoid any toxicity of CAR- expressing cells. See, e.g., Song et al. Cancer Gene Ther. 2008; 15(10):667-75; Clinical Trial Id. No. NCT02107963; and Di et al. N. Engl. J. Med. 2011; 365:1673-83.
- Alternative strategies for regulating the CAR therapy include utilizing small molecules or antibodies that deactivate or turn off CAR activity, e.g., by deleting CAR-expressing cells, e.g., by inducing antibody dependent cell-mediated cytotoxicity (ADCC).
- CAR-expressing cells described herein may also express an antigen that is recognized by molecules capable of inducing cell death, e.g., ADCC or compliment-induced cell death.
- CAR expressing cells described herein may also express a receptor capable of being targeted by an antibody or antibody fragment.
- receptors examples include EpCAM, VEGFR, integrins (e.g., integrins anb3, a4, aI3/4b3, a4b7, a5b1, anb3, an), members of the TNF receptor superfamily (e.g., TRAIL-R1, TRAIL -R2), PDGF Receptor, interferon receptor, folate receptor, GPNMB, ICAM-1, HLA-DR, CEA, CA-125, MUC1, TAG-72, IL-6 receptor, 5T4, GD2, GD3, CD2, CD3, CD4, CD5, CD11, CDlla/LFA-1, CD15, CD18/ITGB2, CD19, CD20, CD22, CD23/lgE Receptor, CD25, CD28, CD30, CD33, CD38, CD40, CD41, CD44, CD51, CD52, CD62L, CD74, CD80, CD125, CD147/basigin, CD152/CTLA-4, CD154/CD40L, CD195
- CAR-expressing cells described herein may also express a truncated epidermal growth factor receptor (EGFR) which lacks signaling capacity but retains the epitope that is recognized by molecules capable of inducing ADCC, e.g., cetuximab (ERBITUX®), such that administration of cetuximab induces ADCC and subsequent depletion of the CAR- expressing cells (see, e.g., WO2011/056894, and Jonnalagadda et al, “ Gene Ther. 2013; 20(8)853- 860).
- EGFR epidermal growth factor receptor
- the CAR cell comprises a polynucleotide encoding a suicide polypeptide, such as for example RQR8. See, e.g., WO2013153391A, which is hereby incorporated by reference in its entirety.
- the suicide polypeptide may be expressed at the surface of a CAR cell.
- the suicide polypeptide may also comprise a signal peptide at the amino terminus.
- Another strategy includes expressing a highly compact marker/suicide gene that combines target epitopes from both CD32 and CD20 antigens in the CAR-expressing cells described herein, which binds rituximab, resulting in selective depletion of the CAR-expressing cells, e.g., by ADCC (see, e.g., Philip et al, Blood 2014; 124(8)1277-1287).
- Other methods for depleting CAR-expressing cells include administration of CAMPATH®, a monoclonal anti-CD52 antibody that selectively binds and targets mature lymphocytes, e.g., CAR-expressing cells, for destruction, e.g., by inducing ADCC.
- the CAR-expressing cell can be selectively targeted using a CAR ligand, e.g., an anti-idiotypic antibody.
- the anti-idiotypic antibody can cause effector cell activity, e.g., ADCC or ADC activities, thereby reducing the number of CAR- expressing cells.
- the CAR ligand, e.g., the anti-idiotypic antibody can be coupled to an agent that induces cell killing, e.g., a toxin, thereby reducing the number of CAR- expressing cells.
- the CAR molecules themselves can be configured such that the activity can be regulated, e.g., turned on and off, as described below.
- a regulatable CAR where the CAR activity can be controlled is desirable to optimize the safety and efficacy of a CAR therapy.
- a RCAR comprises a set of polypeptides, typically two in the simplest embodiments, in which the components of a standard CAR described herein, e.g., an AB domain and an ICS domain, are partitioned on separate polypeptides or members.
- the set of polypeptides include a dimerization switch that, upon the presence of a dimerization molecule, can couple the polypeptides to one another, e.g., can couple an AB domain to an ICS domain. Additional description and exemplary configurations of such regulatable CARs are provided herein and in International Publication No. WO 2015/090229, hereby incorporated by reference in its entirety.
- an RCAR comprises two polypeptides or members: 1) an intracellular signaling member comprising an ICS domain, e.g., a primary ICS domain described herein, and a first switch domain; 2) an antigen binding member comprising an AB domain, e.g., that specifically binds a target molecule described herein, as described herein and a second switch domain.
- the RCAR comprises a TM domain described herein.
- a TM domain can be disposed on the intracellular signaling member, on the antigen binding member, or on both. Unless otherwise indicated, when members or elements of an RCAR are described herein, the order can be as provided, but other orders are included as well.
- the order is as set out in the text, but in other embodiments, the order can be different.
- the order of elements on one side of a transmembrane region can be different from the example, e.g., the placement of a switch domain relative to an ICS domain can be different, e.g., reversed.
- the CAR expressing immune cell may only transiently express a CAR.
- the cells may be transduced with mRNA comprising a nucleic acid sequence encoding an inventive CAR.
- the present disclosure also includes an RNA construct that can be directly transfected into a cell.
- a method for generating mRNA for use in transfection involves in vitro transcription (IVT) of a template with specially designed primers, followed by polyA addition, to produce a construct containing 3' and 5' untranslated sequences ("UTRs"), a 5' cap and/or Internal Ribosome Entry Site (IRES), the nucleic acid to be expressed, and a polyA tail, typically 50-2000 bases in length (SEQ ID NO: 405).
- RNA so produced can efficiently transfect different kinds of cells.
- the template includes sequences for the CAR.
- an RNA CAR vector is transduced into a cell by electroporation.
- the CAR expressing cells may further comprise one or more CARs, in addition to the first CAR. These additional CARs may or may not be specific for the target molecule of the first CAR. In some embodiments, the one or more additional CARs may act as inhibitory or activating CARs. In some aspects, the CAR of some embodiments is the stimulatory or activating CAR; in other aspects, it is the costimulatory CAR. In some embodiments, the cells further include inhibitory CARs (iCARs, see Fedorov et al, Sci. Transl.
- the AB domain of the CAR is or is part of an immunoconjugate, in which the AB domain is conjugated to one or more heterologous molecule(s), such as, but not limited to, a cytotoxic agent, an imaging agent, a detectable moiety, a multimerization domain, or other heterologous molecule.
- Cytotoxic agents include, but are not limited to, radioactive isotopes (e.g., At211, 1131, 1125, Y90, Rel86, Rel88, Sml53, Bi212, P32, Pb212 and radioactive isotopes of Lu); chemotherapeutic agents; growth inhibitory agents; enzymes and fragments thereof such as nucleolytic enzymes; antibiotics; toxins such as small molecule toxins or enzymatically active toxins.
- radioactive isotopes e.g., At211, 1131, 1125, Y90, Rel86, Rel88, Sml53, Bi212, P32, Pb212 and radioactive isotopes of Lu
- chemotherapeutic agents e.g., At211, 1131, 1125, Y90, Rel86, Rel88, Sml53, Bi212, P32, Pb212 and radioactive isotopes of Lu
- growth inhibitory agents e.g., enzymes and
- the AB domain is conjugated to one or more cytotoxic agents, such as chemotherapeutic agents or drugs, growth inhibitory agents, toxins (e.g., protein toxins, enzymatically active toxins of bacterial, fungal, plant, or animal origin, or fragments thereof), or radioactive isotopes.
- cytotoxic agents such as chemotherapeutic agents or drugs, growth inhibitory agents, toxins (e.g., protein toxins, enzymatically active toxins of bacterial, fungal, plant, or animal origin, or fragments thereof), or radioactive isotopes.
- the cells may be further modified to overexpress pro-survival signals, reverse anti-survival signals, overexpress Bcl-xL, overexpress hTERT, lack Fas, or express a TGF-b dominant negative receptor.
- Persistence may also be facilitated by the administration of cytokines, e.g., IF-2, IF-7, and IF-15.
- the present disclosure contemplates the preparation and isolation of a clonal population of antigen-specific B-cells that may be used for isolating at least one CoV-S antigen-specific cell, which can be used to produce a monoclonal antibody against CoV-S, which is specific to a desired CoV-S antigen, or a nucleic acid sequence corresponding to such an antibody.
- Methods of preparing and isolating said clonal population of antigen-specific B-cells are taught, for example, in U.S. Patent Publication No. US2007/0269868 to Carvalho-Jensen et al, the disclosure of which is herein incorporated by reference in its entirety.
- Methods of preparing and isolating said clonal population of antigen-specific B-cells are also taught herein in the examples.
- Methods of “enriching” a cell population by size or density are known in the art. See, e.g., U.S. Patent No. 5,627,052. These steps can be used in addition to enriching the cell population by antigen-specificity.
- the present disclosure contemplates methods for producing anti-CoV- S antibodies and fragments thereof.
- Methods of producing antibodies are well known to those of ordinary skill in the art.
- methods of producing chimeric antibodies are now well known in the art (See, for example, U.S. Patent No. 4,816,567 to Cabilly et al. ⁇ , Morrison et al, Proc. Natl. Acad. Sci. U.S.A., 81:8651-55 (1984); Neuberger et al., Nature, 314:268-270 (1985); Boulianne, G.F.
- Antibody polypeptides of the disclosure having CoV-S binding specificity may also be produced by constructing, using conventional techniques well known to those of ordinary skill in the art, an expression vector containing a promoter (optionally as a component of a eukaryotic or prokaryotic operon) and a DNA sequence encoding an antibody heavy chain in which the DNA sequence encoding the CDRs required for antibody specificity is derived from a non-human cell source, e.g., a rabbit or rodent B-cell source, while the DNA sequence encoding the remaining parts of the antibody chain is derived from a human cell source.
- a promoter optionally as a component of a eukaryotic or prokaryotic operon
- a DNA sequence encoding an antibody heavy chain in which the DNA sequence encoding the CDRs required for antibody specificity is derived from a non-human cell source, e.g., a rabbit or rodent B-cell source, while the DNA sequence encoding the remaining parts of the antibody chain is
- a second expression vector is produced using the same conventional means well known to those of ordinary skill in the art, said expression vector containing a promoter (optionally as a component of a eukaryotic or prokaryotic operon) and a DNA sequence encoding an antibody light chain in which the DNA sequence encoding the CDRs required for antibody specificity is derived from a non-human cell source, e.g., a rabbit or rodent B-cell source, while the DNA sequence encoding the remaining parts of the antibody chain is derived from a human cell source.
- a promoter optionally as a component of a eukaryotic or prokaryotic operon
- a DNA sequence encoding an antibody light chain in which the DNA sequence encoding the CDRs required for antibody specificity is derived from a non-human cell source, e.g., a rabbit or rodent B-cell source, while the DNA sequence encoding the remaining parts of the antibody chain is derived from a human cell source.
- the expression vectors are transfected into a host cell by convention techniques well known to those of ordinary skill in the art to produce a transfected host cell, said transfected host cell cultured by conventional techniques well known to those of ordinary skill in the art to produce said antibody polypeptides.
- the host cell may be co-transfected with the two expression vectors described above, the first expression vector containing DNA encoding a promoter (optionally as a component of a eukaryotic or prokaryotic operon) and a light chain-derived polypeptide and the second vector containing DNA encoding a promoter (optionally as a component of a eukaryotic or prokaryotic operon) and a heavy chain-derived polypeptide.
- the two vectors contain different selectable markers, but preferably achieve substantially equal expression of the heavy and light chain polypeptides.
- a single vector may be used, the vector including DNA encoding both the heavy and light chain polypeptides.
- the coding sequences for the heavy and light chains may comprise cDNA, genomic DNA, or both.
- the host cells used to express the antibody polypeptides may be either a bacterial cell such as E. coli, or a eukaryotic cell such as P. pastoris.
- a mammalian cell of a well- defined type for this purpose such as a myeloma cell, a CHO cell line, a NSO cell line, or a HEK293 cell line may be used.
- the general methods by which the vectors may be constructed, transfection methods required to produce the host cell and culturing methods required to produce the antibody polypeptides from said host cells all include conventional techniques.
- the cell line used to produce the antibody is a mammalian cell line, any other suitable cell line, such as a bacterial cell line such as an E. coli- derived bacterial strain, or a yeast cell line, may alternatively be used.
- the antibody polypeptides may be purified according to standard procedures in the art, such as for example cross-flow filtration, ammonium sulphate precipitation, affinity column chromatography, hydrophobic interaction chromatography (“HIC”), and the like.
- the antibody polypeptides described herein may also be used for the design and synthesis of either peptide or non-peptide mimetics that would be useful for the same therapeutic applications as the antibody polypeptides of the disclosure (See, for example, Saragobi et ai, Science, 253:792-795 (1991), the contents of which are herein incorporated by reference in its entirety).
- the present disclosure contemplates methods for humanizing antibody heavy and light chains which bind to CoV-S.
- Exemplary methods for humanizing antibody heavy and light chains that may be applied to anti-CoV-S antibodies are identified herein and are conventional in the art.
- the screening assays described here may be used to identify high affinity anti-CoV-S Abs which may be useful in the treatment of diseases and disorders associated with CoV-S in subjects exhibiting symptoms of a CoV-S associated disease or disorder.
- the antibody is used as a diagnostic tool.
- the antibody can be used to assay the amount of CoV-S present in a sample and/or subject.
- such antibodies need not be neutralizing antibodies.
- the diagnostic antibody is not a neutralizing antibody.
- the diagnostic antibody binds to a different epitope than the neutralizing antibody binds to.
- the two antibodies do not compete with one another.
- the antibodies disclosed herein are used or provided in an assay kit and/or method for the detection of CoV-S in mammalian tissues or cells in order to screen/diagnose for a disease or disorder associated with changes in levels of CoV-S.
- the kit comprises an antibody that binds CoV-S and means for indicating the binding of the antibody with CoV-S, if present, and optionally CoV-S protein levels.
- Various means for indicating the presence of an antibody can be used. For example, fluorophores, other molecular probes, or enzymes can be linked to the antibody and the presence of the antibody can be observed in a variety of ways.
- the method for screening for such disorders can involve the use of the kit, or simply the use of one of the disclosed antibodies and the determination of whether the antibody binds to CoV-S in a sample.
- high or elevated levels of CoV-S will result in larger amounts of the antibody binding to CoV-S in the sample.
- degree of antibody binding can be used to determine how much CoV-S is in a sample.
- Subjects or samples with an amount of CoV-S that is greater than a predetermined amount e.g., an amount or range that a person without a CoV-S -related disorder would have
- the present disclosure further provides for a kit for detecting binding of an anti-CoV-S antibody of the disclosure to CoV-S.
- the kit may be used to detect the presence of CoV- S specifically reactive with an anti-CoV-S antibody or an immunoreactive fragment thereof.
- the kit may also include an antibody bound to a substrate, a secondary antibody reactive with the antigen and a reagent for detecting a reaction of the secondary antibody with the antigen.
- a kit may be an ELISA kit and can comprise the substrate, primary and secondary antibodies when appropriate, and any other necessary reagents such as detectable moieties, enzyme substrates, and color reagents, for example as described herein.
- the diagnostic kit may also be in the form of an immunoblot kit.
- the diagnostic kit may also be in the form of a chemiluminescent kit (Meso Scale Discovery,
- the diagnostic kit may also be a lanthanide -based detection kit (PerkinElmer, San Jose, CA).
- a biological sample includes, but is not limited to, sera, plasma, urine, fecal sample, saliva, mucous, pleural fluid, synovial fluid, and spinal fluid.
- anti-CoV-S antibodies described herein, or antigen-binding fragments thereof are useful for ameliorating or reducing the symptoms of, or treating, or preventing, diseases and disorders associated with CoV-S.
- Anti-CoV-S antibodies described herein, or antigen binding fragments thereof, as well as combinations, can also be administered in a therapeutically effective amount to patients in need of treatment of diseases and disorders associated with CoV-S in the form of a pharmaceutical composition as described in greater detail below.
- Symptoms of CoV infection may include fever, cough, runny nose, congestion, sore throat, bronchitis, pneumonia, shortness of breath, chest pain, headache, muscle ache, chills, fatigue, conjunctivitis, diarrhea, loss of smell, and loss of taste.
- Complications and/or diseases/disorders associated with coronavirus infection may include, for example, bronchitis, pneumonia, respiratory failure, acute respiratory failure, organ failure, multi-organ system failure, pediatric inflammatory multisystem syndrome, acute respiratory distress syndrome (a severe lung condition that causes low oxygen in the blood and organs), blood clots, cardiac conditions, myocardial injury, myocarditis, heart failure, cardiac arrest, acute myocardial infarction, dysrhythmias, venous thromboembolism, post intensive care syndrome, shock, anaphylactic shock, cytokine release syndrome, septic shock, disseminated intravascular coagulation, ischemic stroke, intracerebral hemorrhage, microangiopathic thrombosis, psychosis, seizure, nonconvulsive status epilepticus, traumatic brain injury, stroke, anoxic brain injury, encephalitis, posterior reversible leukoencephalopathy, necrotizing encephalopathy, post- infectious encephalitis
- Certain diseases and conditions such as high blood pressure, type 1 diabetes, liver disease, overweight, chronic lung diseases including cystic fibrosis, pulmonary fibrosis, and asthma, compromised immune system due to transplant, use of an immunosuppressant, or HIV infection, and brain and nervous system condition, may increase the risk of CoV infection-associated complications and diseases.
- the subject anti-CoV-S antibodies and antigen-binding fragments may be used alone or in conjunction with other active agents, e.g., opioids and non-opioid analgesics such as NSAIDs to elicit analgesia.
- other active agents e.g., opioids and non-opioid analgesics such as NSAIDs to elicit analgesia.
- aspirin and/or acetaminophen may be taken in conjunction with the subject anti-CoV-S antibody or antigen-binding fragment.
- Aspirin is another type of non steroidal anti-inflammatory compound.
- the subject antibodies potentially optionally may be combined with one or more of the following: (i) an antiviral drug, optionally, remdesivir, favipiravir, darunavir, nelfinavir, saquinavir, lopinavir, or ritonavir; (ii) an antihelminth drug, optionally ivermectin; (iii) an antiparasitic drug, optionally hydroxychloroquine, chloroquine, or atovaquone; (iv) antibacterial vaccine, optionally the tuberculosis vaccine BCG; or (v) an anti-inflammatory drug, optionally a steroid such as ciclesonide, a TNF inhibitor (e.g., adalimumab), a TNF receptor inhibitor (e.g., etanercept), an IL-6 inhibitor (e.g., clazakizumab), an IL-6 receptor inhibitor (e.g., toclizuma
- the subject to which the pharmaceutical formulation is administered can be, e.g., any human or non-human animal needing such treatment, prevention and/or amelioration, or who would otherwise benefit from the inhibition or attenuation of CoV-S-mediated activity.
- the subject can be an individual that is diagnosed with, or who is deemed to be at risk of being afflicted by any of the aforementioned diseases or disorders.
- the subject may be in an advanced state of CoV infection, e.g., a subject who is on a ventilator.
- the subject can be one having one or more risk factors (such as advanced age, obesity, diabetes, etc, and others previously identified) which correlate to a poor CoV treatment or recovery prognosis.
- the present disclosure further includes the use of any of the pharmaceutical formulations disclosed herein in the manufacture of a medicament for the treatment, prevention and/or amelioration of any disease or disorder associated with CoV or CoV-S activity (including any of the above-mentioned exemplary diseases, disorders and conditions).
- the present invention also provides a method for treating a coronavirus infection in a subject in need thereof, the method comprising administering to the subject an isolated antibody, or antigen binding fragment thereof, wherein the antibody, or antigen-binding fragment thereof, comprises a heavy chain variable region (VH) comprising a VH CDR1 comprising SEQ ID NO:52, a VH CDR2 comprising SEQ ID NO:54, and a VH CDR3 comprising SEQ ID NO:56, and a light chain variable region (VL) comprising a VL CDR1 comprising SEQ ID NO:252, a VL CDR2 comprising SEQ ID NO:254, and a VL CDR3 comprising SEQ ID NO:256; and wherein the antibody, or antigen-binding fragment thereof, is administered at a dosage of at least 300 mg within 5, 4, 3, 2, or 1 days of symptom onset.
- VH heavy chain variable region
- VL light chain variable region
- the antibody, or antigen-binding fragment thereof is administered at a dosage of about 300 mg. In some embodiments, the antibody, or antigen-binding fragment thereof, is administered at a dosage of about 500 mg. In some embodiments, the antibody, or antigen-binding fragment thereof, is administered at a dosage of about 600 mg. In some embodiments, the antibody, or antigen-binding fragment thereof, is administered at a dosage of about 1200 mg. In some embodiments, the antibody, or antigen-binding fragment thereof, is administered at a dosage of about 4500 mg.
- the subject has at least one risk factor which renders them more prone to a poor clinical outcome.
- the at least one risk factor is selected from the group consisting of: an old age selected from the group consisting of over 55, over 60 or over 65 years old; diabetes, a chronic respiratory condition, obesity, hypertension, a cardiac or cardiovascular condition, a chronic inflammatory or autoimmune condition, and an immune compromised status.
- the subject is immunocompromised.
- the subject is at a high risk of disease progression.
- the subject is (a) age 50 years or above with no comorbid conditions or additional risk factors for progression of COVID-19; (b) between about 18 to about 50 years old and with one or more preexisting medical conditions selected from the group consisting of obesity, diabetes, chronic kidney disease, chronic lung disease, cardiac disease, sickle cell disease or thalassemia, solid organ or blood stem cell transplant recipient, other immunodeficiency due to underlying illness or immunosuppressant medication, Down Syndrome, stroke or cerebrovascular disease, substance use disorder, and pregnancy; or (c) between about 12 to about 17 years old and with one or more preexisting medical conditions selected from the group consisting of obesity, diabetes, chronic kidney disease, sickle cell disease or thalassemia, congenital or acquired heart disease, neurodevelopmental disorder, a medically-related technological dependence, asthma or chornic respiratory disease, solid organ or blood stem cell transplant recipient, other immunodeficiency due to underlying illness or immunosuppressant medication, Down Syndrome, stroke or cerebrovascular disease, substance use disorder, and
- the subject is age 50 or above with no comorbid conditions or additional risk factors for progression of COVID-19. In some embodiments, the subject has hypertension with at least one medication prescribed or recommended. In some embodiments, the subject has moderate to severe asthma requiring daily therapy.
- the anti-CoV-S antibodies described herein, or CoV-S binding fragments thereof, as well as combinations of said antibodies or antigen-binding fragments thereof are administered to a subject at a concentration of between 0.1 mg/ml and about any one of 0.5, 1, 5, 10, 15 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 110, 120, 130, 140, 150, 160, 170,
- the anti-CoV-S antibodies and fragments thereof described herein are administered to a subject at a dose of between about 0.01 and 100.0 or 200.0 mg/kg of body weight of the recipient subject.
- about 1 ⁇ g/kg to 50 mg/kg (e.g., 0.1-20 mg/kg) of antibody is an initial candidate dosage for administration to the patient, whether, for example, by one or more separate administrations, or by continuous infusion.
- about 1 ⁇ g/kg to 15 mg/kg (e.g., 0.1 mg/kg- 10 mg/kg) of antibody is an initial candidate dosage for administration to the patient.
- a typical daily dosage might range from about 1 ⁇ g/kg to 100 mg/kg or more, depending on several factors, e.g., the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the agent, the method of administration, the scheduling of administration, and other factors known to medical practitioners. However, other dosage regimens may be useful.
- the subject anti- CoV-S antibodies and antigen-binding fragments thereof can be administered to a subject at an absolute dose (mg). Accordingly, in one embodiment, the anti-CoV-S antibodies and antigen-binding fragments thereof described herein are administered to a subject at a dose of between about 1 microgram and about 2000 milligrams regardless of the route of administration.
- the antibody, or antigen-binding fragment thereof is administered at a dose of about 100 mg to about 5000 mg, about 100 mg to about 4500 mg, about 100 mg to about 4000 mg, about 100 mg to about 3500 mg, about 100 mg to about 3000 mg, about 100 mg to about 2500 mg, about 300 mg to about 4500 mg, about 500 mg to about 4500 mg, about 600 mg to about 4500 mg, about 1200 mg to about 4500 mg, about 100 mg to about 2000 mg, about 200 mg to about 1500 mg, about 300 mg to about 600 mg, about 500 mg to about 1200 mg, or about 300 mg to about 1200 mg.
- the antibody, or antigen-binding fragment thereof is administered at a dose of about 150 mg, about 200 mg, about 300 mg, about 400 mg, about 450 mg, about 500 mg, about 600 mg, about 700 mg, about 800 mg, about 900 mg, about 1000 mg about 1100 mg, about 1200 mg, about 1300 mg, about 1400 mg, about 1500 mg, about 1600 mg, about 1700 mg, about 1800 mg, about 1900 mg, about 2000 mg, about 2500 mg, about 3000 mg, about 3500 mg, about 4000 mg, about 4500 mg, or about 5000 mg.
- the antibody, or antigen-binding fragment thereof is administered intravenously. In other embodiments, the antibody, or antigen-binding fragment thereof, is administered intramuscularly.
- the antibody, or antigen-binding fragment thereof is administered at a dose of about 150 mg intramuscularly, about 300 mg intramuscularly, about 450 mg intramuscularly, about 500 mg intravenously, about 600 mg intramuscularly, about 1200 mg intramuscularly, or about 1200 mg intravenously.
- the antibody, or antigen-binding fragment thereof is administered. In some embodiments, at least two of the antibody, or antigen-binding fragment thereof, are administered.
- the anti-CoV-S antibody and antigen-binding fragment thereof e.g., ADI-58125
- the anti-CoV-S antibody and antigen-binding fragment thereof may be used in combination with a second antibody, or antigen-binding fragment thereof, wherein the second antibody, or antigen-binding fragment thereof, is selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, ADI-58131, or a combination thereof.
- the second antibody, or antigen-binding fragment thereof is ADI-58122. In one embodiment, the second antibody, or antigen-binding fragment thereof, is ADI-58127. In one embodiment, the second antibody, or antigen-binding fragment thereof, is ADI-58129. In one embodiment, the second antibody, or antigen-binding fragment thereof, is ADI-58131.
- the antibody, or antigen-binding fragment thereof is administered once. In one embodiment, the antibody, or antigen-binding fragment thereof, is administered weekly. In another embodiment, the antibody, or antigen-binding fragment thereof, is administered daily, weekly, every two weeks, monthly, or every two months. In one embodiment, the antibody, or antigen-binding fragment thereof, is administered weekly for about four weeks, once weekly for about a month, weekly for about 5 weeks, weekly for about 6 weeks, weekly for about 7 weeks, or weekly for about two months.
- the methods further comprise obtaining a serum sample from the subject.
- the antibody, or antigen-binding fragment thereof reaches a maximum concentration (Cmax) of about 10 ⁇ g/mL to about 1000 ⁇ g/mL, about 20 ⁇ g/mL to about 500 ⁇ g/mL, about 30 ⁇ g/mL to about 400 ⁇ g/mL, about 40 ⁇ g/mL to about 300 ⁇ g/mL, about 50 ⁇ g/mL to about 200 mg/mL, about 50 mg/mL to about 100 mg/mL, about 30 mg/mL to about 70 mg/mL, about 100 mg.mL to about 400 mg/mL, or about 150 mg/mL to about 350 mg/mL in the serum sample of the subject.
- Cmax maximum concentration
- the antibody, or antigen-binding fragment thereof reaches a maximum concentration (Cmax) of about 30 mg/mL, about 40 mg/mL, about 50 mg/mL, about 60 mg/mL, about 70 mg/mL, about 80 mg/mL, about 90 mg/mL, about 100 mg/mL, about 110 mg/mL, about 120 mg/mL, about 130 mg/mL, about 140 mg/mL, about 150 mg/mL, about 160 mg/mL, about 170 mg/mL, about 180 mg/mL, about 190 mg/mL,or about 200 mg/mL, about 210 mg/mL, about 220 mg/mL, about 230 mg/mL, about 240 mg/mL, about 250 mg/mL, about 260 mg/mL, about 270 mg/mL, about 280 mg/mL, about 290 mg/mL, about 300 mg/mL, about 350 mg/mL, or about 400 mg/mL in the serum sample of the
- the median time for the antibody, or antigen-binding fragment thereof, to reach a maximum concentration in the serum sample of the subject is about 5-30 days, about 6-20 days, about 7-18 days, or about 8-15 days, or about 13-15 days after administration. In some embodiments, the median time for the antibody, or antigen-binding fragment thereof, to reach a maximum concentration in the serum sample of the subject is about 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 days after administration.
- the antibody, or antigen-binding fragment thereof reaches a maximum concentration (Cmax) of about 30 ⁇ g/mL to about 100 ⁇ g/mL, about 40 ⁇ g/mL to about 80 ⁇ g/mL, about 50 ⁇ g/mL to about 70 ⁇ g/mL, or about 30 ⁇ g/mL to about 65 ⁇ g/mL in the serum sample of the subject in about 6-20 days, about 7-18 days, about 8-15 or about 13-15 days after administration.
- Cmax maximum concentration
- the area under the serum concentration-time curve from day 0 to day 21 is about 100-2000 day* ⁇ g/mL, about 1000-2000 day* ⁇ g/mL, about 1400-2000 day* ⁇ g/mL, about 200-1500 day* ⁇ g/mL, about 400-1400 day* ⁇ g/mL, about 500-1300 day* ⁇ g/mL, about 600-1000 day* ⁇ g/mL, or about 800-900 day* ⁇ g/mL.
- the area under the serum concentration-time curve from day 0 to day 21 is about 500 day* ⁇ g/mL, about 600 day* ⁇ g/mL, about 700 day* ⁇ g/mL, about 800 day* ⁇ g/mL, about 900 day* ⁇ g/mL, about 1000 day* ⁇ g/mL, about 1100 day* ⁇ g/mL, about 1200 day* ⁇ g/mL, about 1300 day* ⁇ g/mL, about 1400 day* ⁇ g/mL, about 1500 day* ⁇ g/mL, about 1600 day* ⁇ g/mL, about 1700 day* ⁇ g/mL, about 1800 day* ⁇ g/mL, about 1900 day* ⁇ g/mL, or about 2000 day* ⁇ g/mL.
- the area under the serum concentration-time curve from day 0 to day 90 is about 1000-10000 day* ⁇ g/mL, about 2000-5000 day* ⁇ g/mL, about 3000-4000 day* ⁇ g/mL, about 5000-10000 day* ⁇ g/mL, about 5000-8000 day* ⁇ g/mL, or about 6000-8000 day* ⁇ g/mL.
- the area under the serum concentration-time curve from day 0 to day 90 is about 1000 day* ⁇ g/mL, about 2000 day* ⁇ g/mL, about 3000 day* ⁇ g/mL, about 4000 day* mg/mL, about 5000 day* mg/mL, about 6000 day* mg/mL, about 7000 day* mg/mL, about 8000 day* mg/mL, about 9000 day* mg/mL, or about 10000 day* mg/mL.
- the area under the serum concentration-time curve from day 0 to day 180 is about 1000-10000 day* mg/mL, about 2000-5000 day* mg/mL, about 3000-6000 day* mg/mL, about 5000-10000 day* mg/mL, about 5000-8000 day* mg/mL, or about 4000-6000 day* mg/mL.
- the area under the serum concentration-time curve from day 0 to day 180 is about 1000 day* mg/mL, about 2000 day* mg/mL, about 3000 day* mg/mL, about 4000 day* mg/mL, about 5000 day* mg/mL, about 6000 day* mg/mL, about 7000 day* mg/mL, about 8000 day* mg/mL, about 9000 day* mg/mL, or about 10000 day* mg/mL.
- the area under the serum concentration-time curve from day 0 to day 365 is about 1000-30000 day* mg/mL, about 5000-30000 day* mg/mL, about 10000- 30000 day* mg/mL, about 10000-15000 day* mg/mL, about 20000-30000 day* mg/mL, or about 25000-30000 day* mg/mL.
- the area under the serum concentration-time curve from day 0 to day 365 is about 1000 day* mg/mL, about 2000 day* mg/mL, about 3000 day* mg/mL, about 4000 day* mg/mL, about 5000 day* mg/mL, about 6000 day* mg/mL, about 7000 day* mg/mL, about 8000 day* mg/mL, about 9000 day* mg/mL, about 10000 day* mg/mL, about 11000 day* mg/mL, about 12000 day* mg/mL, about 13000 day* mg/mL, about 14000 day* mg/mL, about 15000 day* mg/mL, about 20000 day* mg/mL, about 21000 day* mg/mL, about 22000 day* mg/mL, about 23000 day* mg/mL, about 24000 day* mg/mL, about 25000 day* mg/mL, about 26000 day* mg/mL, about
- the antibody, or antigen-binding fragment thereof has a serum half- life of about 50-200 days, about 50-160 days, about 50-140 days, about 40-130 days, about 50-100 days, about 60-100 days, about 60-90 days, about 70-90 days, about 90-100 days, about 70-80 days, about 80-130 days, about 100-150 days, about 110-140 days, about 120-140 days.
- the antibody, or antigen-binding fragment thereof has a serum half-life of about 50, 55, 60, 65, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117, 118,
- the method further comprises obtaining an epithelial lining fluid (ELF) sample from the subject.
- ELF sample may comprise an ELF sample from the upper airway, e.g., nasopharyngeal or oropharyngeal; the lower airway, e.g., lung, and/or an alveolar tissue.
- the antibody, or antigen-binding fragment thereof reaches a concentration of about 1 mg/mL to about 100 mg/mL, about 1 mg/mL to about 80 mg/mL, about 80 mg/mL to about 100 mg/mL, about 50 mg/mL to about 100 mg/mL, about 1 mg/mL to about 50 mg/mL, about 2 mg/mL to about 25 mg/mL, or about 2 mg/mL to about 10 mg/mL in the ELF sample of the subject.
- the antibody, or antigen-binding ragment thereof has a receptor occupancy of at least 50%, 60%, 70%, 80%, or 90% in the ELF sample.
- Receptor occupany can be linked to a viral dynamic model to enable the prediction of the natural time course of viral load and the effect of the antibody, or antigen-binding fragment thereof, on viral clearance and infectivity rate.
- RO can be calculated using: 1) in vitro SARS-CoV-2 binding kinetics of the antibody, or antigen-binding fragment thereof, e.g., the association rate constant (k on ) and the dissociation rate constant (k 0ff ) , e.g., obtained from a Biacore assay; 2) time course of the concentrations of the antibody, or antigen-binding fragment thereof, in a sample, e.g., an ELF sample; and 3) time course of viral load following administration of the antibody, or antigen-binding fragment thereof.
- association rate constant k on
- k 0ff dissociation rate constant
- administration of the antibody, or antigen-binding fragment thereof results in at least 50%, 60%, 70%, 80%, or 90% SARS-CoV-2 receptor occupancy in the ELF sample. In some embodiments, administration of the antibody, or antigen-binding fragment thereof, results in a durable, e.g., about 20-day, about 25-day, about 28-day, about 30-day, about 40-day, about 50-day, about 60-day, SARS-CoV-2 receptor occupancy in the ELF sample.
- administration of the antibody, or antigen-binding fragment thereof maintains the concentration of the antibody, or antigen binding fragment thereof, above a concentration (e.g., about 0.27 mg/L or about 0.5 mg/L) associated with about 95% or 100% viral growth suppression in an in vitro post infection assay.
- a concentration e.g., about 0.27 mg/L or about 0.5 mg/L
- intramuscular (IM) administration of ADI-58125 at a dose of about 300 mg results in about 90% SARS-CoV-2 receptor occupacy in the ELF sample for at least 28 days, and maintains the concentration of ADI-58125 in the ELF sample above the concentration associated with 100% viral growth suppression in vitro, i.e., 0.5 mg/mL.
- intramuscular (IM) administration of ADI-58125 at a dose of about 300 mg results in about 90% SARS-CoV-2 receptor occupacy in the ELF sample for at least 28 days, and maintains the concentration of ADI-58125 in the ELF sample above the concentration associated with 95% viral growth suppression in vitro, i.e., 0.274 mg/mL.
- administering results in a 50% virus neutralization titer (MN50) of about 100-2000, e.g., within the range of peak serum virus-neutralizing antibody (sVNA) titers for COVID-19 vaccine recipients.
- MN50 virus neutralization titer
- sVNA peak serum virus-neutralizing antibody
- administration of the antibody, or antigen-binding fragment thereof maintains a serum concentration of the antibody, or antigen-binding fragment thereof, about 100-fold higher than the in vitro 90% inhibitory concentration (IC 90 ) of 0.011 ⁇ g/mL against authentic SARS-CoV-2 (USA- WA1) for a minimum of 6 months.
- IC 90 in vitro 90% inhibitory concentration
- the virus neutralization titer of the antibody, or antigen-binding fragment thereof is determined using a plaque reduction neutralization test (PRNT), as described in Example 1.
- PRNT plaque reduction neutralization test
- the PRNT is a serological test which utilizes the ability of a specific antibody to neutralize a virus, in turn, preventing the virus from causing the formation of plaques in a cell monolayer (Stephen J. et al., The American Journal of Tropical Medicine and Hygiene. 81 (5): 825- 833).
- the assay involves mixing a constant amount of virus with dilutions of the serum specimen being tested, followed by plating of the mixture onto cells of an appropriate cell line for the individual virus.
- the concentration of plaque forming units can be determined by the number of plaques formed after a few days.
- a vital dye e.g., neutral red
- the plaque forming units are measured by microscopic observations, fluorescent antibodies, or specific dyes that react with the infected cell.
- the PRNT test is considered the "gold standard" for detecting and measuring antibodies that can neutralize the viruses that cause many diseases. It has a higher sensitivity and is more specific than many other serological tests for the diagnosis of some viruses.
- the 80% neutralization titer is reported using a validated method, e.g., SOP-VC-M204.
- the 50% neutralization titer is reported.
- the antibody, or antigen-binding fragment thereof has a virus neutralizing titer of about 100-2000, about 200-1500, about 300-1500, or about 500-1500 in the serum sample of the subject about 3, 6, or 12 months after administration. In some embodiments, the antibody, or antigen-binding fragment thereof, has a virus neutralizing titer of about 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, 1100, 1200, 1300, 1400, 1500, 1600, 1700, 1800, 1900 or 2000 in the serum sample of the subject about 3, 6 or 12 months after administration.
- the 80% virus neutralization titer (MN80) of the antibody, or antigen binding fragment thereof, at about days 0-14, about days 7-21, about 3 months, or about 6 months after administration is about 10-6000, about 50-600, about 500-1500, about 1000-2500, about 100- 2500, about 500-2000, about 500-1500, about 400-1200, about 200-1500, about 300-1000, about 400- 800, about 400-1000, or about 500-600 in the subject.
- the 80% virus neutralization titer (MN80) of the antibody, or antigen binding fragment thereof, at about day 7, day 14, or day 21 after administration is about 300-2000, about 400-600, about 600-1500, about 1100-1700, about 500-1700, about 500-1500, about 400-1200, about 400-800, about 400-1000, or about 500-600 in the serum sample of the subject.
- the 80% virus neutralization titer (MN80) of the antibody, or antigen binding fragment thereof, at about 3 months after administration is about 200-1000, about 200-800, about 200-500, about 400-900, or about 400-600 in the serum sample of the subject.
- the 80% virus neutralization titer (MN80) of the antibody, or antigen binding fragment thereof, at about 6 months after administration is about 10-500, about 300-500, or about 50-200 in the serum sample of the subject.
- the 50% virus neutralization titer (MN50) of the antibody, or antigen binding fragment thereof, at about days 0-14, about days 7-21, about 3 months, about 6 months, or about 12 months after administration is about 100-6000, about 300-1500, about 1700-3800, about 3800-5200, about 300-5500, about 1200-4500, about 1300-4300, about 1200-4000, about 100-2500, about 500-2500, about 800-2000, about 1000-1800, about 800-1300, about 900-1100, or about 1300- 1500 in the serum sample of the subject.
- the 50% virus neutralization titer (MN50) of the antibody, or antigen binding fragment thereof, at about day 7, day 14 or day 21 after administration is about 1000-4500, about 1200-4500, about 1300-4300, about 1200-3900, about 1500-4000, about 1800-3800, about 3800-4500, about 1000-1800 or about 1200-1500 in the serum sample of the subject.
- the 50% virus neutralization titer (MN50) of the antibody, or antigen binding fragment thereof, at about 3 months after administration is about 800-1300 or about 900- 1100 in the serum sample of the subject. In some embodiments, the 50% virus neutralization titer (MN50) of the antibody, or antigen-binding fragment thereof, at about 6 months after administration, is about 200-500 or about 300-600 in the serum sample of the subject.
- the 50% virus neutralization titer (MN50) of the antibody, or antigen binding fragment thereof, at about 12 months after administration is about 150-500 or about 200-400 in the serum sample of the subject.
- the serum concentration of the antibody, or antigen-binding fragment thereof is about 1-300 mg/L, about 1-250 mg/L, about 1-200 mg/L, about 1-100 mg/L, about 100-250 mg/L, about 150-200 mg/L, about 120-170 mg/L, about 1-90 mg/L, about 1-80 mg/L, about 1-70 mg/L, about 1-60 mg/L, about 1-50 mg/L, about 1-40 mg/L, about 1-30 mg/L, about 1-20 mg/L, about 1-10 mg/L, about 2-9 mg/L, about 3-8 mg/L, about 4-6 mg/L, about 5-80 mg/L, about 10-70 mg/L, or about 10-60 mg/L in the subject about 3 months after administration.
- the serum concentration of the antibody, or antigen-binding fragment thereof is about 1 mg/L, about 2 mg/L, about 3 mg/L, about 4 mg/L, about 5 mg/L, about 6 mg/L, about 7 mg/L, about 8 mg/L, about 9 mg/L, about 10 mg/L, about 11 mg/L, about 12 mg/L, about 13 mg/L, about 14 mg/L, about 15 mg/L, about 16 mg/L, about 17 mg/L, about 18 mg/L, about 19 mg/L, about 20 mg/L, about 25 mg/L, about 30 mg/L, about 35 mg/L, about 40 mg/L, about 45 mg/L, about 50 mg/L, about 55 mg/L, about 60 mg/L, about 65 mg/L, about 70 mg/L, about 75 mg/L, about 80 mg/L, about 85 mg/L, about 90 mg/L, about 95 mg/L, about 100 mg/L, about 110 mg/L, about 120 mg/L, about
- the serum concentration of the antibody, or antigen-binding fragment thereof is about 1-100 mg/L, about 1-90 mg/L, about 1-80 mg/L, about 1-70 mg/L, about 1-60 mg/L, about 1-50 mg/L, about 1-40 mg/L, about 1-30 mg/L, about 1-20 mg/L, about 1-10 mg/L, about 2-9 mg/L, about 3-8 mg/L, about 4-6 mg/L, about 5-80 mg/L, about 10-70 mg/L, about 10-60 mg/L, about 5-25 mg/L, about 10-30 mg/L, about 15-25 mg/L, about 20-100 mg/L, about 30-100 mg/L, about 50- 80 mg/L, or about 60-70 mg/L in the subject about 6 months after administration.
- the serum concentration of the antibody, or antigen-binding fragment thereof is about 1 mg/L, about 2 mg/L, about 3 mg/L, about 4 mg/L, about 5 mg/L, about 6 mg/L, about 7 mg/L, about 8 mg/L, about 9 mg/L, about 10 mg/L, about 11 mg/L, about 12 mg/L, about 13 mg/L, about 14 mg/L, about 15 mg/L, about 16 mg/L, about 17 mg/L, about 18 mg/L, about 19 mg/L, about 20 mg/L, about 25 mg/L, about 30 mg/L, about 35 mg/L, about 40 mg/L, about 45 mg/L, about 50 mg/L, about 55 mg/L, about 60 mg/L, about 65 mg/L, about 70 mg/L, about 75 mg/L, about 80 mg/L, about 85 mg/L, about 90 mg/L, about 95 mg/L, or about 100 mg/L in the subject about 6 months after administration.
- the serum concentration of the antibody, or antigen-binding fragment thereof is about 0.1-30 mg/L, about 1-30 mg/L, about 1-20 mg/L, about 1-10 mg/L, about 10-20 mg/L, about 0.1-3 mg/L, about 0.5-8 mg/L, about 0.5-10 mg/L, about 2-9 mg/L, about 5-15 mg/L, about 3-8 mg/L, or about 4-6 mg/L in the subject about 12 months after administration.
- the serum concentration of the antibody, or antigen-binding fragment thereof is about 0.1 mg/L, about 0.2 mg/L, about 0.3 mg/L, about 0.4 mg/L, about 0.5 mg/L, about 0.6 mg/L, about 0.7 mg/L, about 0.8 mg/L, about 0.9 mg/L, about 1 mg/L, about 2 mg/L, about 3 mg/L, about 4 mg/L, about 5 mg/L, about 6 mg/L, about 7 mg/L, about 8 mg/L, about 9 mg/L, about 10 mg/L, about 15 mg/L, about 20 mg/L, about 25 mg/L, or about 30 mg/L in the subject about 12 months after administration.
- the clearance rate for the antibody, or antigen-binding fragment thereof is about 0.1-10 mL/d, about 0.1-5.0 mL/d, about 0.1-3.0 mL/d, about 0.5-2.5 mL/d, or about 1.0-2.0 mL/d.
- the steady state volume of distribution for the antibody, or antigen binding fragment thereof is about 1-10 L, about 2-8 L, about 4-9 L, about 4-8 L, or about 5-7L.
- administration of the antibody, or antigen-binding fragment thereof reduces the risk of COVID-19 hospitalization or death of the subject.
- the risk of COVID-19 hospitalization or death is reduced by at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90% or 95%.
- administration of the antibody, or antigen-binding fragment thereof reduces viral load in the subject.
- the viral load is reduced by at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90% or 95%.
- the viral load is reduced by at least about 0.5 loglO copies/mL, at least about 0.6 loglO copies/mL, at least about 0.7 loglO copies/mL, at least about 0.8 loglO copies/mL, at least about 0.9 loglO copies/mL, at least about 1.0 loglO copies/mL, at least about 1.1 loglO copies/mL, at least about 1.2 loglO copies/mL, at least about 1.3 loglO copies/mL, at least about 1.4 loglO copies/mL, at least about 1.5 loglO copies/mL, at least about 1.6 loglO copies/mL, at least about 1.7 loglO copies/mL, at least about 1.8 loglO copies/mL, at least about 1.9 loglO copies/mL, at least about 2.0 loglO copies/mL, at least about 2.1 loglO copies/mL, at least about 2.2 loglO copies/mL, at least about 2.3 loglO copies/mL
- the anti-CoV-S antibodies described herein, or anti-CoV-S antigen-binding fragments thereof, as well as combinations of said antibodies or antigen-binding fragments thereof are administered to a recipient subject with a frequency of once every twenty-six weeks or less, such as once every sixteen weeks or less, once every eight weeks or less, once every four weeks or less, once every two weeks or less, once every week or less, or once daily or less.
- the antibody containing medicament or pharmaceutical composition is peripherally administered to a subject via a route selected from one or more of: orally, sublingually, buccally, topically, rectally, via inhalation, transdermally, subcutaneously, intravenously, intra-arterially, or intramuscularly, via intracardiac administration, intraosseously, intradermally, intraperitoneally, transmucosally, vaginally, intravitreally, epicutaneously, intra- articularly, peri-articularly, or locally.
- Fab fragments may be administered every two weeks or less, every week or less, once daily or less, multiple times per day, and/or every few hours.
- a patient receives Fab fragments of 0.1 mg/kg to 40 mg/kg per day given in divided doses of 1 to 6 times a day, or in a continuous perfusion form, effective to obtain desired results.
- the concentration of the antibody or Fab administered to a given patient may be greater or lower than the exemplary administration concentrations set forth above.
- a person of skill in the art would be able to determine an effective dosage and frequency of administration through routine experimentation, for example guided by the disclosure herein and the teachings in, Goodman & Gilman's The Pharmacological Basis of Therapeutics, Brunton, L.L. et al. editors, 11 th edition, New York, New York: McGraw-Hill (2006); Howland, R. D. et al, Pharmacology, Volume 864, Lippincott's illustrated reviews., Philadelphia, PA: Lippincott Williams & Wilkins (2006); and Golan, D. E., Principles of pharmacology: the pathophysiologic basis of drug therapy, Philadelphia, PA: Lippincott Williams & Wilkins (2007).
- the anti-CoV-S antibodies described herein, or CoV-S binding fragments thereof, as well as combinations of said antibodies or antigen-binding fragments thereof are administered to a subject in a pharmaceutical formulation.
- the subject is a human.
- the subject is an adult. In some embodiment, the subject is an adolescent. In some embodiments, the subject is a child or a pediatric subject. In one embodiment, the subject is a pediatric subject, e.g., from birth to age of 11 years old, e.g., birth to ⁇ 1 month, 1 month to ⁇ 2 years, or 2-11 years old. In other embodiments, the adolescent is 12-17 years old or 12- 15 years old. In one embodiment, the adult is over the age of 18. In some embodiments, the subject has no known recent exposure to COVID-19. In some embodiments, the subject has a known recent exposure to COVID-19.
- recent exposure is within the last 14 days, within the last 10 days, within the last 7 days, within the last 6 days, 5 days, 4 days, 3 days, 2 days, or 24 hours.
- the subject has COVID-19 symptoms or an active COVID-19 infection.
- the subject has had COVID-19 symptoms for less than 5 days, less than 4 days, less than 3 days, less than 2 days, or less than 1 day.
- the subject is treated within 5 days, 4 days, 3 days, 2 days or 1 day of symptom onset.
- early treatment e.g., receiving treatment within 5 days, 4, days, 3 days, 2 days or 1 day of symptom onset, provides a reduction in the risk of COVID-19 related hospitalization and all-cause death.
- the subject e.g., the adult, the adolescent, or the child
- the subject is vaccinated or has received a COVID-19 vacination.
- the subject e.g., the adult, the adolescent, or the child
- the subject e.g., the adult, the adolescent, or the child, is at risk of COVID-19 exposure.
- the subject may be a first responder, doctor, nurse, clinican, etc.
- the subject is immunocompromised.
- the subject is a significantly immune compromised subject, e.g., adult subject or pediatric subject, but whose underlying medical condition(s) or treatments to treat those conditions cause to the subjects to be immunocompromised, and/or put them at increased risk of developing severe symptomatic COVID- 19.
- the subject has no known recent exposure to COVID-19.
- the subject has a known recent exposure to COVID-19.
- recent exposure is within the last 14 days, within the last 10 days, within the last 7 days, within the last 6 days, 5 days, 4 days, 3 days, 2 days, or 24 hours.
- the immunocomprised subject includes, but is not limited to, an individual who is actively treated for solid tumor or hematologic malignancies; solid organ transplant (SOT) recipient taking immunosuppressive therapy; CAR-T-cell therapy or HCT recipient; subjects having moderate or severe primary immunodeficiency; subjects having advanced or untreated HIV infection, or who are taking high-dose corticosteroids, alkylating agents, antimetabolites, or TNF blockers.
- the immunocompromised subject is vaccinated or has received a COVID-19 vacination. In some embodiments, the immunocompromised subject is not vaccinated or has not received a COVID-19 vacination.
- the subject is at a high risk of COVID progression.
- the subjects at a high risk of disease progression are age 50 years old or above with no comorbid conditions or additional risk factors for progression of COVID-19.
- the subjects at a high risk of disease progression are those about 18 to about 50 years old and with one or more preexisting medical conditons selected from the group consisting of (i) obesity (body mass index (BMI) > 30 kg/m 2 ), (ii) diabetes (type 1 or type 2); (iii) chronic kidney disease (eGFR calculated by modification of diet in renal disease (MDRD) of 59 mL/min/1.73 m 2 or less, including end-stage renal disease on hemodialysis); (iv) chronic lung disease (emphysema/chronic obstructive pulmonary disease, chronic bronchitis, interstitial lung disease [including idiopathic pulmonary fibrosis], cystic fibrosis, or moderate to severe asthma [defined as requiring daily therapy]); (v) cardiac disease (heart failure, coronary artery disease, cardiomyopathies, or hypertension [with at least one medication prescribed or recommended]); (vi) sickle cell disease or thalassemia
- the subjects at a high risk of disease progression are those about 12 to about 17 years old (inclusive) and with one or more preexisting medical conditions selected from the group consisting of (i) BMI >85th percentile for age and sex based on United States Center for Disease Control (CDC) growth charts; (ii) diabetes (Type 1 or Type 2); (iii) chronic kidney disease; (iv) sickle cell disease or thalassemia; (v) congenital or acquired heart disease; (vi) neurodevelopmental disorders (e.g., cerebral palsy, Down syndrome); (vii) a medically-related technological dependence (e.g., tracheostomy, gastrostomy, or positive pressure ventilation not related to COVID-19); (viii) asthma, reactive airway or other chronic respiratory disease that requires daily medication for control; (ix) solid organ or blood stem cell transplant recipients; (x) other immunodeficiency due to underlying illness or immunosuppressant medication; (xi) substance use disorder; or (x
- the subject is age 50 or above with no comorbid conditions or additional risk factors for progression of COVID-19.
- the subject has hypertension with at least one medication prescribed or recommended.
- the subject has moderate to severe asthma requiring daily therapy.
- a “pharmaceutical composition” or “medicament” refers to a chemical or biological composition suitable for administration to a subject, preferably a mammal, more preferably a human.
- compositions may be specifically formulated for administration via one or more of a number of routes, including but not limited to buccal, epicutaneous, epidural, inhalation, intraarterial, intracardial, intracerebroventricular, intradermal, intramuscular, intranasal, intraocular, intraperitoneal, intraspinal, intrathecal, intravenous, oral, parenteral, rectally via an enema or suppository, subcutaneous, subdermal, sublingual, transdermal, and transmucosal.
- routes including but not limited to buccal, epicutaneous, epidural, inhalation, intraarterial, intracardial, intracerebroventricular, intradermal, intramuscular, intranasal, intraocular, intraperitoneal, intraspinal, intrathecal, intravenous, oral, parenteral, rectally via an enema or suppository, subcutaneous, subdermal, sublingual, transdermal, and transmucosal.
- administration can occur
- the anti-CoV-S antibodies or antigen-binding fragments thereof, as well as combinations of said antibodies or antigen-binding fragments thereof, may be optionally administered in combination with one or more active agents.
- active agents include (i) an antiviral drug, optionally, remdesivir, favipiravir, darunavir, nelfinavir, saquinavir, lopinavir, or ritonavir; (ii) an antihelminth drug, optionally ivermectin; (iii) an antiparasitic drug, optionally hydroxychloroquine, chloroquine, or atovaquone; (iv) antibacterial vaccine, optionally the tuberculosis vaccine BCG; or (v) an anti-inflammatory drug, optionally a steroid such as ciclesonide, a TNF inhibitor (e.g., adalimumab), a TNF receptor inhibitor (e.g., e
- An anti -histamine can be any compound that opposes the action of histamine or its release from cells (e.g., mast cells).
- Anti-histamines include but are not limited to acrivastine, astemizole, azatadine, azelastine, betatastine, brompheniramine, buclizine, cetirizine, cetirizine analogues, chlorpheniramine, clemastine, CS 560, cyproheptadine, desloratadine, dexchlorpheniramine, ebastine, epinastine, fexofenadine, HSR 609, hydroxyzine, levocabastine, loratadine, methscopolamine, mizolastine, norastemizole, phenindamine, promethazine, pyrilamine, terfenadine, and tranilast.
- antibiotics include but are not limited to amikacin, aminoglycosides, amoxicillin, ampicillin, ansamycins, arsphenamine, azithromycin, azlocillin, aztreonam, bacitracin, carbacephem, carbapenems, carbenicillin, cefaclor, cefadroxil, cefalexin, cefalothin, cefalotin, cefamandole, cefazolin, cefdinir, cefditoren, cefepime, cefixime, cefoperazone, cefotaxime, cefoxitin, cefpodoxime, cefprozil, ceftazidime, ceftibuten, ceftizoxime, ceftobiprole, ceftriaxone, cefuroxime, cephalosporins, chloram
- Active agents also include aldosterone, beclomethasone, betamethasone, corticosteroids, cortisol, cortisone acetate, deoxycorticosterone acetate, dexamethasone, fludrocortisone acetate, glucocorticoids, hydrocortisone, methylprednisolone, prednisolone, prednisone, steroids, and triamcinolone. Any suitable combination of these active agents is also contemplated.
- a “pharmaceutical excipient” or a “pharmaceutically acceptable excipient” is a carrier, usually a liquid, in which an active therapeutic agent is formulated.
- the active therapeutic agent is a humanized antibody described herein, or one or more fragments thereof.
- the excipient generally does not provide any pharmacological activity to the formulation, though it may provide chemical and/or biological stability, and release characteristics. Exemplary formulations can be found, for example, in Remington’s Pharmaceutical Sciences, Gennaro, A. editor, 19 th edition, Philadelphia, PA: Williams and Wilkins (1995), which is incorporated by reference.
- pharmaceutically acceptable carrier includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic, and absorption delaying agents that are physiologically compatible.
- the carrier is suitable for parenteral administration.
- the carrier can be suitable for intravenous, intraperitoneal, intramuscular, or sublingual administration.
- Pharmaceutically acceptable carriers include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. The use of such media and agents for pharmaceutically active substances is well known in the art.
- compositions typically must be sterile and stable under the conditions of manufacture and storage.
- the disclosure contemplates that the pharmaceutical composition is present in lyophilized form.
- the composition can be formulated as a solution, microemulsion, liposome, or other ordered structure suitable to high drug concentration.
- the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol), and suitable mixtures thereof.
- the disclosure further contemplates the inclusion of a stabilizer in the pharmaceutical composition.
- the proper fluidity can be maintained, for example, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
- isotonic agents for example, sugars, polyalcohols such as mannitol and sorbitol, or sodium chloride in the composition.
- Absorption of the injectable compositions can be prolonged by including an agent that delays absorption, for example, monostearate salts and gelatin.
- the alkaline polypeptide can be formulated in a time-release formulation, for example in a composition that includes a slow release polymer.
- the active compounds can be prepared with carriers that will protect the compound against rapid release, such as a controlled release formulation, including implants and microencapsulated delivery systems.
- Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, poly anhydrides, polyglycolic acid, collagen, polyorthoesters, polylactic acid, polylactic and polyglycolic copolymers (“PLG”). Many methods for the preparation of such formulations are known to those skilled in the art. [0585] For each of the recited embodiments, the compounds can be administered by a variety of dosage forms. Any biologically acceptable dosage form known to persons of ordinary skill in the art, and combinations thereof, are contemplated.
- dosage forms include, without limitation, reconstitutable powders, elixirs, liquids, solutions, suspensions, emulsions, powders, granules, particles, microparticles, dispersible granules, cachets, inhalants, aerosol inhalants, patches, particle inhalants, implants, depot implants, injectables (including subcutaneous, intramuscular, intravenous, and intradermal), infusions, and combinations thereof.
- Example 1 Phase 1, Randomized, Double-blind, Single Ascending Dose Study [0590] This Example provides safety, pharmacokinetics, and virus neutralizing antibody titer data of ADI-58125 in a Phase 1, randomized, double-blinded, single ascending dose study. In this Phase 1 study, three dose levels of ADI-58125: 300 mg IM, 500 mg IV, and 600 mg IM, were assessed in healthy participants. All dose levels utilized a 100 mg/ml concentration of ADI-58125.
- One participant from Cohort 1 (300 mg IM) reported a single episode of dizziness, which occurred shortly after IM injection and resolved after the participant was permitted to eat. This AE was graded mild, deemed unrelated to study medication, and resolved without concomitant medication administration (see, Tables 11 and 12).
- One participant in Cohort 2 (500 mg IV) reported musculoskeletal chest pain beginning approximately 10 hours after infusion and resolving prior to discharge the following morning. No abnormalities were found on physical exam or on an ECG performed during the event. This event was graded mild, deemed unrelated to study medication, and resolved without intervention or concomitant medication administration.
- a second participant in Cohort 2 (500 mg IV) reported a single episode of vomiting. This event occurred approximately 24 hours after infusion and resolved immediately after the event and prior to discharge. No abnormalities were found on physical exam shortly after the event. This event was graded mild, deemed unrelated to study medication, and resolved without intervention or concomitant medication administration.
- One participant in Cohort 3 (600 mg IM) reported bruising on their left thigh, which they noticed incidentally 11 days after their injection and reported at her Day 14 visit. This finding was not noted during injection site monitoring, which included physical exams, nor was it noted or reported at their Day 7 visit. This event was graded mild, deemed unrelated to study medication.
- AE Adverse event
- TEAE Treatment emergent adverse event
- SAE Serious adverse event
- N is number of participants in the Safety Population. Percentages are based on the total number of participants in Safety [0600] Population.
- TEAE is defined as any event not present prior to the initiation of study drug or any event already present that worsens in either intensity or frequency following exposure to study drug.
- AE counts For participant counts, if participants experienced one or more events, they are counted only once. For AE counts, each occurrence of an AE is counted. AEs are summarized at maximum Severity and strongest Relationship to study drug.
- Adverse event terms are coded using MedDRA version 23.1.
- TEAE Treatment emergent adverse event.
- N number of participants in the Safety Population. Percentages are based on the total number of participants in Safety Population.
- Solicited AEs are local reactions to the IM injection and are collected through Study Day 4 and are imputed as related to study drug.
- TEAEs is defined as any event not present prior to the initiation of study drug or any event already present that worsens in either intensity or frequency following exposure to study drug.
- Adverse event terms are coded using MedDRA version 23.1.
- PK Pharmacokinetics
- T max The median time to maximum concentration
- AUC area under the serum concentration-time curve
- Cmax maximum concentration
- SD standard deviation
- Tmax time to maximum concentration
- T 1/2 half life.
- VNA virus neutralizing antibody
- peak live virus ID50 geometric mean VNA titers for the Moderna vaccine mRNA- 1273 were -1000 for all age groups tested at Day 43 post vaccination 1 and decayed to 406, 171 and 131 by Day 209 in participants aged 18 to 55, 56 to 70 and 71 years of age or older, respectively (Doria-Rose N, et al, N Engl J Med. 2021 Apr 6). Taken together, these data support the idea that lower VNA titers are protective against symptomatic disease.
- Serum virus neutralizing titers were evaluated using blood samples collected prior to dosing (predose) and at 8 h, 24 h (Day 2), Day 7, and Day 14 postdose.
- This value is likely within the range of 50% SARS-CoV-2 neutralizing GMT titers remaining -6 months post vaccination for mRNA vaccines such as BNT162b2 and mRNA-1273 (Wang et al, Nature. 2021 Feb 10:1-7; Doria-Rose N, et al., N Engl J Med. 2021 Apr 6).
- Serum viral neutralizing antibody (sVNA) titers were also assessed Day 2, Day 7, Day 14, Day 21, 3 months, and 6 months post dose in subjects following a single IM administration of 300 mg ADI-58125, a single IM administration of 600 mg ADI-58125, and a single IV administration of 500 mg ADI-58125.
- the sVNA data are presented in Table 14.
- ADI-58125 sVNA titers against authentic SARS-CoV-2 were compared with AZD1222- and mRNA-1273-induced titers measured by plaque reduction neutralization assay.
- ADI-58125 serum concentration was highly predictive of the MN50 sVNA titer ( Figure 20). Table 14. Neutralizing Antibody Titers by Study Day and Treatment Groups
- IM intramuscular
- IV intravenous
- MN50 50% neutralization titer
- MN80 80% neutralization titer.
- MN50 sVNA titers associated with ADI-58125 were significantly higher than pean titers associated with AZD1222 (P ⁇ 0.0001) and mRNA-1273 (P ⁇ 0.01), and were maintained at comparable levels to titers associated with mRNA-1273 through Study Month 6 ( Figure 18), suggesting that the ADI-58125 neutralizing antibody titers also support potential for prolonged protection.
- sVNA titers following ADI-58125 administration were also determined by plaque reduction assay for SARS-CoV-2 BavPat (D614G), B.1.351/Beta and the B.1.617.2/Delta variants. These were compared with titers 7-30 days after the second dose of authorized vaccines in a separate cohort of volunteers. As shown in Figure 27, 80% neutralization (MN80) sVNA titres following a single 300 mg IM injection of ADI-58125 exceeded titers against each of the BavPat, B.1.351/Beta and B.1.617.2/Delta variants 7-30 days following 2-dose mRNA-1273 vaccination, and this trend was maintained for at least 3 months.
- MN80 neutralization
- Example 2 Serum Titers of ADI-58125 Against SARS-CoV-2 Variants by Plaque Reduction Neutralization Test
- Plaque reduction neutralization test was used to measure serum titers against SARS- CoV-2 D614G and B.1.351 variants. Briefly, 400 PFU SARS-CoV-2 virus was incubated with serial dilutions of ADI-58125 for 1 hour. Virus/antibody mixtures were then added to a confluent monolayer of Vero-E6 cells and incubated for 48 hours before adding neutral red agar overlay. Plaques were counted 24 hours after infection and the concentration of antibody that reduced the plaques by 50% (PRNT50) or 90% (PRNT90) relative to control (i.e., virus alone with no antibody) was calculated (Figure 4).
- Example 3 Post-Infection Assay with ADI-58125 Against SARS-CoV-2 Variants
- a post-infection assay was performed to measure the concentration of antibody required to prevent viral replication of the SARS-CoV-2 D614G and B.1.351 variants in infected cells. Briefly, cells were infected with the SARS-CoV-2 D614G or B.1.351 variants. After 1 hour incubation, unbound virus was washed away, and medium containing various concentration of ADI-58125 antibody was added to infected-cell monolayers. Viral supernatant was sampled daily and infectious viral burden was quantified by plaque assay on Vero E6 cells ( Figure 5).
- C.37/Lambda variant, and the Mu/B.1621 variant were assessed.
- SARS-CoV-2 antibodies were included as comparators. Briefly, neutralizing activity against authentic SARS-CoV-2 virus varaints was assessed in a focus reduction neutralization test using Vero cells.
- Half-maximal inhibitory concentration (IC 50 ) values were determined using nonlinear regression curve fitting.
- SARS-CoV-2 variants containing single or double amino acid mutations in the spike protein and full sets of mutations observed in variants of interest and variants of concern were assessed in a lentivial pseudovirus assay.
- D614G an early variant of SARS-CoV-2, was used as a reference to calculate the IC 50 fold change in neutralizing activity of the antibodies.
- In vitro neutralizing activity of ADI-58125 against SARS-CoV-2 variants, including those reported to exhibit reduced susceptibility to emergency-use authorized (EUA) mAbs was assessed in a non-replicative vesicular stomatitis virus pseudovirus assay. Fold reduction was calculated by dividing the IC50 of a variant by the mean IC50 (14.6 ng/mL) of the D614G reference strain.
- ADI-58125 and ADI-58122 were able to neutralize all common circulating SARS-CoV-2 variants and emerging lineages.
- ADI- 58125 retained in vitro neutralizing activity within 0.4- to 5.1-fold relative to the reference D614G variant for all pseudovirus variants tested, including the newly emerging lineages including the Alpha/B.1.1.7 variant, the Beta/B.1.351 variant, the Gamma/P.l variant, the B.1.617 (e.g.,
- IC50 values for ADI- 58125 (within 0.6- to 2.4-fold of the D614G strain) were lower than those observed for bamlanivimab, etesevimab, casirivimab, imdevimab, and sotrovimab. These data support further clinical investigation of ADI-58125 not only for COVID-19 but also for potential utility in future outbreaks caused by other SARS-like viruses.
- Example 5 A Whole-Body Quantitative System Pharmacology Physiologically-Based Pharmacokinetic (QSP/PBPK) Model that a priori Predicts Intramuscular (IM) Pharmacokinetics of ADI-58125: an Extended Half-life Monoclonal Antibody for the Treatment and Prevention of Coronavirus Disease ( COVID-19 )
- This Example provides a QSP/PBPK model, which was constructed using ADI-58125- specific physiochemical properties and published non-human primate (NHP) and human PK data for other antibodies, to a priori predict and confirm NHP and human PK.
- NHP non-human primate
- An existing QSP/PBPK model was modified to include 3 distinct lung sub-compartments: upper airway, lower airway, and alveolar tissue (Figure 9A). Each sub-compartment ( Figure 9B) contained an epithelial lining fluid (ELF) space ( Figure 9B).
- the model was fit separately to digitized NHP and human serum PK data for 7 extended half-life antibodies to estimate the apparent neonatal Fc receptor (FcRn) binding affinity and bioavailability by drug.
- FcRn neonatal Fc receptor
- Nasopharyngeal swab (upper airway) and lung (lower airway) ELF PK data from 4 additional antibodies were used to optimize a single rate constant for transcytosis in lung.
- NHP ADI-58125 K D,FcRn was optimized to be 35.7 nM and human ADI-58125 K D,FcRn (9.55 nM) was derived using a mean NHP:human K D,FcRn ratio of 3.74 across antibodies.
- Model-based simulated human serum PK data using inter-subject variability from NHP and actual weight distribution from an ongoing Phase 1 study aligned with initial 21 -day data (Figure 9D). Using an adult CDC weight distribution (45-150 kg), the simulated median half-life was 74.4 days.
- the modified QSP/PBPK model was optimized by estimating NHP intramuscular (IM) bioavailability and ADI-58125 K D,FcRn and applying an NHP:human K D,FcRn ratio to the NHP K D,FcRn values estimated for ADI-58125 to better forecast human ADI-58125 concentration-time profiles.
- IM intramuscular
- PI prediction interval
- Figures 22A-22B show the initial QSP/PBPK model-forecasted NHP median (90% prediction interval (PI)) serum ADI-58125 concentration-time profile following a single intravenous (IV) and intramuscular (IM) administration of 10 mg/kg ADI-58125 with measured concentration data overlaid.
- Figures 23A-23B show the optimized QSP/PBPK model-forecasted NHP median (90% PI) serum ADI-58125 concentration-time profile following a single intravenous (IV) and intramuscular (IM) administration of 10 mg/kg ADI-58125 with measured concentration data overlaid.
- FIGS. 24A-24D show the observed and optimized QSP/PBPK model-forecasted human median (90% PI) serum ADI-58125 concentration-time profiles in healthy adult participants following a 300 mg IM, 500 mg IM or 500 mg IV administration of ADI-58125 with measured concentration data overlaid.
- the modeling strategy involved the modification of a platform whole-body PBPK model designed for wild-type IgGl mAbs to forecast the PK of an extended half-life mAh.
- the modified QSP/PBPK model account for the altered binding affinity to FcRn and included patches of positive charge (PPC) as a covariate on the rate of pinocytosis into the endosomal space (CLup).
- PPC positive charge
- the model adequately a priori predicted the observed ADI-58125 PK in NHPs and human, thus supporting the selected dose.
- This innovative QSP-based modeling and simulation approach enabled the evaluation of candidate dose regimens prior to the availability of PK data, supporting the rapid advancement of the ADI-58125 clinical program during the COVID-19 pandemic.
- Example 6 A Whole-Body Quantitative System Pharmacology Physiologically-Based Pharmacokinetic (QSP/PBPK) Model to Support Dose Selection of ADI-58125 for the Treatment of Coronavirus Disease (COVID-19)
- This Example provides a QSP/PBPK model to support dose selection for a Phase 2/3 COVID- 19 treatment study of ambulatory patients with mild to moderate COVID-19 (STAMP: NCT04805671).
- the QSP/PBPK model comprised 15 specific tissues and one representing the rest of the body; each tissue was connected through blood and lymph flow to the systemic circulation. In tissue endothelial spaces, mAbs enter by pinocytosis (CL up ) and via the interaction with neonatal Fc receptor (FcRn), FcRn-bound mAh is recycled, and unbound drug is eliminated (K deg ).
- the QSP/PBPK model was modified such that the lung compartment was subdivided into alveoli and upper (naso- /oropharyngeal) and lower lung airway compartments (bronchi; Figure 9A).
- mAbs were also allowed to cross epithelial cells by transcytosis (k trans ) and, upon entering the interstitial space, to either exit the lung via lymph flow or re-ente the vascular space via FcRn-mediated recycling k trans was calibrated using serum PK data along with EFF and nasopharyngeal swab PK data from reference mAbs MHAA4549A, VIS-410, ASN-1, and ASN-2. It was assumed that binding to SARS-CoV-2 virus does not impact the ADI-58125 PK at clinically relevant doses.
- a QSP/PBPK model was used to simulate receptor occupancy (RO) and drug exposure in the upper airway (nasopharyngeal/oropharyngeal epithelial lining fluid [EFF] compartment).
- RO was linked to an existing viral dynamic model to enable the prediction of the natural time course of viral load and the effect of ADI-58125 on viral clearance and infectivity rate.
- RO was calculated using: 1) in vitro ADI-58125-SARS-CoV-2 binding kinetics (association rate constant (k on ) of 1.52E+06 M -1 •s -1 and dissociation rate constant (k 0ff ) of 2.81E-04 s -1 from a Biacore assay; 2) time course of QSP/PBPK model-forecasted ADI-58125 concentrations in upper aiway EFF of lung; and 3) time course of viral load following ADI-58125 administration at peak viral load ( e.g ., log 10 7 to log 10 9 copies/mF for all patients) using in vitro 50% half maximal concentration (IC 50 ) of 0.007 ⁇ g/mF against authentic SARS-CoV-2 (e.g., Delta variant).
- Molar SARS-CoV-2 viral binding site capacity was calculated assuming 40 spike proteins per virion, 3 binding sites per spike, for all patients.
- the QSP/PBPK model and a 2018 CDC reference body weight distribution (45-150 kg) were used to simulate 1000 concentration-time profiles for a range of candidate ADI-58125 regimens.
- ADI- 58125 regimens were evaluated against 2 criteria: 1) ability to attain near complete (>90%), and durable (28-day) SARS-CoV-2 RO in the ELF; and 2) ability to maintain ELF ADI-58125 concentrations >0.27 mg/L, which was associated with 95% viral growth suppression in an in vitro post-infection assay against the Beta variant.
- FIG. 10A shows the QSP model-predicted median (90% prediction interval, PI) ADI-58125 PK profiles associated with viral growth suppression. Times to attaining relevant target concentrations are provided below.
- Table 15 shows ADI-58125 potency against SARS-CoV-2 variants of concern.
- ADGI-58125 As shown in Figure 10A and Table 15, ADGI-58125 attained ELF concentration above 10 times the IC90 of other variants of concern.
- Figure 10B shows the ADI-58125 receptor occupancy at various Delta variant virion densities after a single 300 mg IM injection of ADI-58125.
- ADI-58125 300 mg IM dose for the treatment of COVID-19.
- ADI-58125 is forecasted to attain near complete (>90%) SARS-CoV-2 RO in the ELF and maintain ELF ADI-58125 concentrations above that associated with 95% viral growth suppression in vitro.
- the 300 mg IM regimen has a projected ability to rapidly achieve and maintain target tissue concentrations at key tissue sites of viral replication, including the ability to attain near complete (>90%) and durable (28-day) SARS-Cov-2 across a range of baseline viral loads.
- Example 7 A Whole-Body Quantitative System Pharmacology Physiologically-Based Pharmacokinetic (QSP/PBPK) Model to Support Dose Selection of ADI-58125 for the Prevention of Coronavirus Disease (COVID-19)
- This Example provides a QSP/PBPK model to support dose selection for a Phase 2/3 COVID- 19 prevention study (EVADE: NCT04859517).
- the QSP PBPK model forecasts in humans were based upon the estimated IM bioavilability from NHP, while the K D,FcRn value of 9.55 nM was derived based upon multiplying the mean NHP:human K D,FcRn ratio for other extended half-life antibodies to the NHP K D,FcRn value for ADI-581258.
- the QSP/PBPK model was later optimized by estimating K D,FcRn (4.27 nM) and IM bioavailability (92.2%) using the interim human PK data, along with estimating inter-individual variability for some key parameters to better reflect observed variability.
- ADI-58125 IM dosing regimens were evaluated against two criteria: (1) the ability to attain measured 50% neutralization (MN50) serum virus-neutralizing antibody (sVNA) titers within the range of peak sVNA titers for COVID-19 vaccine recipients; and (2) the ability to maintain serum ADI-58125 concentrations 100-fold higher than the in vitro 90% inhibitory concentration (IC90) of 0.011 ⁇ g/mL against authentic SARS-CoV-2 (USA-WA1) for a minimum of 6 months in >90% of simulated patients.
- MN50 50% neutralization
- sVNA serum virus-neutralizing antibody
- IC90 in vitro 90% inhibitory concentration
- This threshold was based on a precedent with respiratory syncytial virus and HIV, in which serum concentrations approximately 100-fold higher than the in vitro IC50 were associated with protection in animal models and/or in humans.
- ADI-58125 maintains MN50 titers within the range of those achieved by COVID-19 vaccine recipients following 2 doses (AZD1222, mean titer 80: mRNA-1273, mean titer 327).
- ADI-58125 52-week post-dose medium serum concentration of 5.3 mg/L and a regression relating ADI- 58125 concentration and MN50 titer, the predicted MN50 is 231 one year post-dose.
- an ADI-58125 dose of > 55 ⁇ g was associated with protection from weight loss compared with controls at Day 6.
- Hamsters receiving the highest dose (2000 ⁇ g) had no detectable virus in lung samples ( Figure 13).
- treatment with 333 ⁇ g and 2000 ⁇ g of ADI-58125 significantly reduced infectious viral loads at Day 3 post exposure compared with controls (P ⁇ 0.005). Similar trends were observed for viral sub-genomic RNA (sgRNA), which was produced during active viral infection.
- sgRNA viral sub-genomic RNA
- Example 9 In vivo Prophylactic Efficacy of ADI-58125 in Non Human Primates
- Viral load was assessed in nasopharyngeal (NP) and oropharyngeal (OP) samples (daily), and in bronchoalveolar lavage (BAL) samples (Days 1, 3, and 5) by reverse transcription-polymerase chain reaction (RT-PCR, all samples) and plaque assays (NP and OP swabs only).
- NP nasopharyngeal
- OP oropharyngeal
- BAL bronchoalveolar lavage
- an ADI-58125 dose of 25 mg/kg was associated with reduced viral replication in the upper and lower airways with sgRNA below the limit of detection in all respiratory compartments tested. Specifically, a similar trend was observed for genomic RNA data (data not shown). Substantial protection was also observed at the 5 mg/kg dose level, as demonstrated by accelerated clearance of infectious virus. In addition, no viral particles were detected in NP and OP compartments at the 25 mg/kg ADI-58125 dose ( Figure 16).
- Example 10 A Phase 1/2, Open-Label, Comparator-Controlled Study Evaluating the Safety, Tolerability, and SARS-CoV-2 Antibody Titers of ADI-58125 Supplementation vs. BNT162b2 COVID-19 Vaccine Boost in Previously Vaccinated adults
- a Phase 1/2, randomized, open label, comparator-controlled, study is conducted in adults with no known history of Coronavirus Disease 19 (COVID-19) or Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection and who are a minimum of 6 months out from completion of a primary BNT162b2 COVID-19 vaccine series at the time of study drug dosing. Participants receive a single open-label IM injection of ADI-58125 or BNT162b2 vaccine boost and will be followed for 12 months.
- COVID-19 Coronavirus Disease 19
- SARS-CoV-2 Severe Acute Respiratory Syndrome Coronavirus 2
- Participants per cohort receive study drug (ADI-58125 or BNT162b2).
- Adult male and female participants must be age >18 years at the time of consent.
- Participants who are healthy or have stable chronic medical conditions, and who are a minimum of 6 months out from completion of a primary COVID-19 vaccine series at the time of study drug dosing are enrolled and screened for participation within 28 days before the scheduled administration of study drug.
- Screening procedures commence after written informed consent is obtained and include confirmation of inclusion and exclusion criteria, medical history, documentation of prior COVID-19 vaccination and medications (i.e., medications taken within 30 days before the scheduled dose of study drug), viral RT-PCR and serology tests, baseline laboratory testing including serum virus neutralizing antibody titers, vital sign measurements, and physical examination.
- Key inclusion criteria include male or female subjects age >18 years at the time of consent; healthy or with stable chronic medical conditions, including immunocompromise, defined as disease not requiring significant change in therapy or hospitalization for worsening disease during the 6 weeks before enrollment; received and completed BNT162b2 COVID-19 vaccine series under EUA or approved dosing guidelines at least 6 months prior to study drug dosing.
- Key inclusion criteria include known history of SARS-CoV-2 infection, prior administration of SARS-CoV vaccine other than the BNT162b2 COVID-19 vaccine series, orior administration of SARS-CoV-2 monoclonal antibody, history of severe adverse reaction associated with a vaccine or monoclonal antibody and/or severe allergic reaction (eg, anaphylaxis) to any component of the study intervention(s), bleeding diathesis of condition associated with prolonged bleeding that would m in the opinion of the investigator, contraindicate intramuscular injection.
- Participants eligible for randomization are admitted to the clinical unit the morning of Day 1 and are confined in the clinical unit for a minimum of 1 hour following study drug administration. Each participant receives a single IM dose of the study drug.
- Serum virus binding and neutralizing antibody titers are assessed at specific timepoints through 12 months.
- Participants are monitored for potential cases of COVID-19. If, at any time, a participant develops acute respiratory illness, the participant can contact the site and an in-person or telehealth visit should occur during which a SARS-CoV-2 RT-PCR test is conducted and serum virus neutralizing antibody titers are measured.
- Secondary endpoints include:
- Exploratory endpoints include incidence of asymptomatic SARS-CoV-2 infection as determined by positive serology to N protein.
- Geometric mean titers (GMT) and the associated 2-sided CIs are derived by calculating means and CIs on the natural log scale based on the t-distribution, and then exponenting the results.
- GMFR and 2-sided 95% CIs are provided for each treatment group at the following timepoints: Days 7, 14 and 28; GMFRs are limited to participants with nomissing values at baseline and the specified post-dose time point.
- the GMFR is calculated as the mean of the difference of logarithmically transformed assay results (later time point - baseline) and transformed back to the original scale.
- Two-sided CIs are obtained by calculating CIs using Student’s t-distribution for the mean difference of the logarithmically transformed assay results and transforming the limits back to the original scale.
- Serum PK parameters are calculated for ADI-58125 using non-compartmental analysis methods. Descriptive statistics (min, max, median, mean, standard deviation, and coefficient of variation) of all PK parameters are provided.
- this study provides evaluation of the magnitude of the SARS-CoV-2 neutralizing/binding antibody response in previously vaccinated adults receiving ADI-58125 supplementation compared to COVID-19 vaccine boost; the safety and tolerability of ADI-58125 supplementation; the pharmacokinetics and the immunogenecity of ADI-58125; and the incidence of RT-PCR confirmed, symptomatic COVID-19 in previously vaccinated adults receiving ADI-58125 supplementation compared to COVID-19 vaccine boost.
- Example 11 Evaluation of ADI-58125 as a Preventive Option in Immunocompromised Individuals
- This Example provides evaluation of ADI-58125 in an immunocompromised population in the prevention of symptomatic COVID-19.
- This cohort includes significantly immune compromised adult participants with no known recent exposure but whose underlying medical condition(s) put them at increased risk of developing severe symptomatic COVID-19.
- Enrolled participants include, for example, subjects actively treated for solid tumor or hematologic malignancies, SOT recipient taking immunosuppressive therapy, CAR-T-cell therapy or HCT recipient, subjects having moderate or severe primary immunodeficiency, subjects having advanced or untreated HIV infection, or subjects currently taking high-dose corticosteroids, alkylating agents, antimetabolites, or TNF blockers.
- Example 12 Evaluation of ADI-58125 as a Treatment or Preventive Option in Pediatric Population
- This Examples provides evaluation of ADI-58125 in pediatric population.
- a separate open- label cohort including children from bith to 11 years of age is added to (1) the phase 2/3 randomized, double-blinded, placebo-controlled trial to evaluate the efficacy and safety of ADI-58125 in the treatment of ambulatory participants with mild or moderate COVID-19 (STAMP), and (2) the phase 2/3 randomized, double blind, placebo controlled trial to evaluate the efficacy and safety of ADI- 58125 in the prevention of COVID-19 (EVADE).
- the dosing regimen can be the below: a) 15 mg/kg for children from birth to ⁇ 1 month b) 10 mg/kg for 1 month to ⁇ 2 years c) 5 mg/kg for 2 to 11 years
- ADI-58125 Children from birth to 11 years of age with symptoms of mild or moderate COVID-19 and a positive SARS-CoV-2 test are enrolled to receive a single IM dose of ADI-58125. Participants are enrolled simultaneously across age groups. ADI-58125 is administered at the study site and participants are monitored after dosing for acute worsening of disease, hypersensitivity, and injection site reactions. After Day 1, participants are monitored through Day 29 for safety, PK, viral titers and clinical outcome. In addition, participants continue long-term follow-up for safety, PK, ADA and development of post-acute sequelae of COVID-19 through approximately 5 half-lives. Prevention study
- the dose regimen can be the below: a) 10 mg/kg for birth to ⁇ 1 month (with a second 10 mg/kg dose 4 months post first dose) b) 10 mg/kg for 1 month to ⁇ 2yrs c) 5 mg/kg for 2 to 11 years
- the preliminary sample size estimations for the planned pediatric studies are based on a predicted CV% of 30% in relevant PK parameters [e.g., area under the concentration-time curve (AUC) and maximum observed concentration (C max )]. It is expected that between 4 and 8 children for each age group will be enrolled to provide > 80% power to obtain reasonably precise estimates (i.e., 95% Cl with 60 and 140% of the geometric mean) of AUC and C max for each age group. To ensure adequate sample size in each age category, PK data for each age group are pooled across the two studies for analysis. It is anticipated that the total sample size includes 16 participants in the 2 to 11 and 1 month to ⁇ 2 year age groups and 8 participants in the birth to ⁇ 1 month age group.
- relevant PK parameters e.g., area under the concentration-time curve (AUC) and maximum observed concentration (C max ). It is expected that between 4 and 8 children for each age group will be enrolled to provide > 80% power to obtain reasonably precise estimates (i.e., 95% Cl with 60 and 14
- ADI-58125 Children from birth to 11 years of age are enrolled to receive a single IM dose of ADI-58125. Participants are enrolled simultaneously across age groups. ADI-58125 will be administered at the study site and participants will be monitored after dosing for acute hypersensitivity and injection site reactions. After Day 1, participants are monitored through approximately 5 half-lives for safety, PK, ADA and the development of SARS-CoV-2 infection (symptomatic or asymptomatic). Participants who develop signs or symptoms consistent with a COVID-like illness (CLI) are evaluated for laboratory confirmed COVID-19. Participants with laboratory confirmed COVID-19 are monitored for clinical and virological outcomes.
- CLI COVID-like illness
- Example 13 Population Pharmacokinetics (PPK) of ADI-58125, an Extended-Half-life Monoclonal Antibody Being Developed for the Treatment and Prevention of COVID-19 [0706]
- This Examples provides a population pharmacokinetics (PPK) model that describes the serum ADI-58125 concentration-time profile following intravenous (IV) and intramuscular (IM) administration in Phase 1 and Phase 2/3 COVID-19 prevention and treatment studies.
- the ADI-58125 PPK model was developed on PK data from a phase 1 first-in-human, single- ascending dose study (24 adults, IV and IM) and from phase 2/3 COVID-10 prevention (EVADE, 659 adults, IM) and treatment (STAMP, 189 adults, IM) studies. 1,486 PK samples were included in the analysis. Data from a phase 1 study were used to develop the base structural model describing the serum disposition of ADI-58125. The base structural model was adapted from a preliminary model developed using data from non-human primate studies. As feasible, the impact of body weight was included in the model, consistent with models for other mAbs.
- the model was qualified using visual predictive check (VPC) plots, and the precision of PPK parameters was evaluated using bootstrap/resampling methods.
- VPC visual predictive check
- the impact of covariates (e.g. body weight, age, and baseline viral load) on ADI-58125 serum disposition were evaluated.
- Prediction-corrected visual predictive check (PC- VPC) plots were used to qualify the PPK model.
- Subject-specific estimates of ADI-58125 exposures were generated using individual, post hoc PK parameters.
- the PPK model comprised 2 systemic compartments, zero-order infusion for IV administration and first-order absorption for IM administration, and provided a robust fit to the data based on the VPC plots (Figure 28) and goodness-of-fit plots (Figure 49).
- the impact of body weight on the variability in total body clearance, distributional clearance, and distributional volume parameters was evaluated and included in the model.
- a fitted exponent of 0.658 was found to be most appropriate for the relationship between body weight and the two clear ane terms, body clearance and distributional clearance, while a direct linear relationship with an exponent of 1.0 was most appropriate for the volume of the central and peripheral compartments.
- Body weight influenced clearance, intercompartmental clearance, and central and peripheral volume compartments.
- the relationship between body weight and clearance was not suggestive of the need for dose adjustment over the population weight range studied (38.6 to 178.7 kg).
- the population mean IM bioavailability estimate was 90.5%.
- Figure 54 shows the PPK model median (90% confidence interval) concentration-time profile following a single 300 mg IM ADI-58125 dose by study.
- the population pharmacokinetic model provides a precise and unbiased fit to observed ADI- 58125 concentration-time in healthy adults.
- the VPC plots shows that model-based simulations recapture the observed data reliable ( Figure 28).
- the traditional goodness-of-fit plots showed that the fit to the data is precise and unbiased both on a population mean and individual basis ( Figure 49).
- the conditional weighted residual plots indicate that there is minimal bias relative to the population fitted concentrations and time since dose.
- AUC area under the curve.
- Cmax maximum serum concentration.
- CL clearance. Tm ⁇ half-life.
- Vss steady state volume of distribution.
- the median population prediction of elimination half-life was 123 days, ranging from 79-277 days. The results are consistent with the intended PK characteristics of ADI-58125 (i.e., prolonged half-life and high IM bioavailability).
- the median AUC from time zero to 6 months and Cams increased in a dose-proportional manner when comparing the 300 mg and 600 mg IM groups. This is consistent with the linear elimination found to be most appropriate during population PK model development.
- Figure 51 shows the population mean predicted concentrations overtime with IV or iM administration. At a dose of 300 mg, ADI-58125 had a robust IM bioavailability. Absorpotion from the IM depot resulted in lower peak concentrations than those seen with the IV depot, but profiles are similar after about 2 months. The population mean estimate of IM bioavailability was 92.2%, which is also confirmed by the population PK model.
- a 2-compartment PPK model with linear elimination and first-order IM absorption provided a precise and unbiased fit to the observed ADI-58125 concentration-time data.
- ADI-58125 demonstrated high IM bioavailability and a median terminal elimination half- life of 125 to 136 days.
- the results are consistent with the intended PK characteristics of ADI-58125 in terms of prolonged half-life and high IM bioavailability.
- This PPK model is useful for future PK- pharmacodynamic analyses and simulations conducted to support phase 2/3 dose selection.
- ADI-58125 can be used in adults and pediatric individuals:
- ADI-58125 may be used in adults and pediatric individuals for post-exposure prophylaxis of
- COVID-19 in individuals who are at high risk for progression to severe COVID-19, including hospitalization or death, and are o not fully vaccinated or o who are not expected to mount an adequate immune response to complete SARS- CoV-2 vaccination (for example, individuals with immunocompromising conditions including those taking immunosuppressive medications) and
- ADI-58125 can be used in pediatric individuals:
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Virology (AREA)
- Medicinal Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Molecular Biology (AREA)
- Immunology (AREA)
- Biophysics (AREA)
- Biochemistry (AREA)
- Genetics & Genomics (AREA)
- Pharmacology & Pharmacy (AREA)
- Animal Behavior & Ethology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Pulmonology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Engineering & Computer Science (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- General Chemical & Material Sciences (AREA)
- Microbiology (AREA)
- Mycology (AREA)
- Epidemiology (AREA)
- Oncology (AREA)
- Communicable Diseases (AREA)
- Peptides Or Proteins (AREA)
- Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
Abstract
The present disclosure is directed to antibodies, and antigen binding fragments thereof, having binding specificity for the S protein of coronaviruses (CoV-S), such as the S protein of the SARS coronavirus (SARS-CoV-S) and/or the S protein of the SARS coronavirus 2 (SARS-CoV-2-S), including neutralizing antibodies and antibodies that bind to and/or compete for binding to the same linear or conformational epitope(s) on CoV-S. Further disclosed are conjugates of anti-CoV-S antibodies, and binding fragments thereof, conjugated to one or more functional or detectable moieties. Methods of making said anti-CoV-S antibodies and antigen binding fragments thereof are also contemplated. Other embodiments of the disclosure include the use of anti-CoV-S antibodies, and binding fragments thereof, for the diagnosis, assessment, and treatment of diseases and disorders associated with coronaviruses, or the S protein thereof, and conditions where neutralization or inhibition of coronaviruses, or the S protein thereof, would be therapeutically and/or prophylactically beneficial.
Description
COMPOUNDS SPECIFIC TO CORONA VIRUS S PROTEIN AND USES THEREOF
RELATED APPLICATIONS
[0001] This application claims the benefit of priority to U.S. Provisional Application 63/182,033, filed April 30, 2021; U.S. Provisional Application No. 63/191,410, filed May 21, 2021; U.S. Provisional Application No. 63/197,717, filed June 7, 2021; U.S. Provisional Application No. 63/211,334, filed June 16, 2021; U.S. Provisional Application No. 63/233,443, filed August 16, 2021; U.S. Provisional Application No. 63/243,259, filed September 13, 2021; U.S. Provisional Application No. 63/245,509, filed September 17, 2021; US Provisional Application No. 63/249,158, filed September 28, 2021; US Provisional Application No. 63/256,753, filed October 18, 2021; US Provisional Application No. 63/279,202, filed November 15, 2021; US Provisional Application No. 63/281,914, filed November 22, 2021; US Provisional Application No. 63/283,433, filed November 27, 2021; U.S. Provisional Application No. 63/292,781, filed December 22, 2021; US Provisional Application No. 63/298,832, filed January 12, 2022; US Provisional Application No. 63/302,714, filed January 25, 2022; US Provisional Application No. 63/316,067, filed March 3, 2022; US Provisional Application No. 63/324,569, filed March 28, 2022; U.S. Provisional Application No. 63/324,995, filed March 29, 2022; U.S. Provisional Application No. 63/327,436, filed April 5, 2022; and U.S. Provisional Application No. 63/332,312, filed April 19, 2022. The entire contents of each of these applications are incorporated herein by reference.
SEQUENCE LISTING
[0002] The instant application contains a Sequence Listing which has been submitted electronically in ASCII format and is hereby incorporated by reference in its entirety. Said ASCII copy, created on April 25, 2022, is named 132280_00420_SL.txt and is 234,693 bytes in size.
FIELD
[0003] This disclosure generally pertains to antibodies and antigen-binding fragments thereof, preferably human antibodies and antigen-binding fragments and/or affinity-matured variants thereof, recombinant cells engineered to express such antibodies, and compositions containing such antibodies and antigen-binding fragments thereof, wherein such antibodies and antigen-binding fragments thereof specifically bind to the S protein of coronaviruses (“CoV-S”) and therapeutic and diagnostic uses for the antibodies, antigen-binding fragments, and compositions thereof.
BACKGROUND
[0004] Coronaviruses (“CoV”) are genetically classified into four major genera: the Alphacoronavirus genus (ACoV genus); the Betacoronavirus genus (BCoV genus); the
Gammacoronavirus genus (CCoV genus); and Deltacoronavirus genus (DCoV genus), and while ACoV and BCoV primarily infect a als CCoV and DCoV predominantly infect birds (Wu A. et al, Cell Host Microbe. 2020 Mar ll;27(3):325-328). Coronaviruses that infect humans were first identified in the mid-1960s, and currently, seven confirmed CoV species are known as human pathogens. Four CoV species, the HCoV-HKUl and HCoV-OC43 from the BCoV genus and the HCoV-229E and HCoV-NL63 from the ACoV genus, are endemic species in humans and cause mild respiratory symptoms, mostly in pediatric patients (Brielle E.S., et al, BioRxiv reprint, doi: https://doi.org/10.1101/2020.03.10.986398). The other three human CoV species, the SARS-CoV, the MERS-CoV, and the SARS-CoV-2 (also known as “2019-nCoV”), all of which are from the BCoV genus, have caused severe outbreaks, including the Severe Acute Respiratory Syndrome (SARS) outbreak in 2002-2003, the Middle East Respiratory Syndrome (MERS) outbreak in 2012-2013, and the current (2019-) pandemic of the coronavirus disease of 2019 (“COVID-19”).
[0005] The genome of coronaviruses, whose size ranges between approximately 26,000 and 32,000 bases, includes a variable number (from 6 to 11) of open reading frames (“ORFs”) (Wu A. et al. , Cell Host Microbe. 2020 Mar ll;27(3):325-328). The first ORF encodes 16 non-structural proteins (“nsps”), and the remaining ORFs encode accessory proteins and structural proteins. The four major structural proteins are the spike surface glycoprotein (“S protein” or “S” or “spike protein”), small envelope protein (“E protein” or “E”), matrix protein (“M protein” or “M”), and nucleocapsid protein (“N protein”, or “N”).
[0006] The S protein, which plays an essential role in binding to receptors on the host cell and determines host tropism (Zhu Z. et al, Infect Genet Evol. 2018 Jul;61 : 183-184), forms homotrimers protruding from the viral surface (Li F. Annu Rev Virol. 2016 Sep 29;3(1):237-261). The S protein is processed into two non-covalently associated subunits, SI and S2, and each monomer in the trimeric S assembly is a heterodimer of SI and S2 subunits. Cryo-EM studies have revealed that the SI subunit is comprised of four domains: an N-terminal domain (NTD), a C-terminal domain (CTD), and two subdomains (Walls A. C. et al, Nature 531, 114-117 (2016).; Tortorici M. A. and Veesler D.,
Adv Virus Res. 2019;105:93-116; Wrapp D. et al, Science 367, 1260-1263 (2020)). The CTD functions as the receptor-binding domain (RBD) for both SARS-CoV and SARS-CoV-2 (Li F. J Virol. 2015 Feb;89(4): 1954-64). The S2 subunit contains the fusion peptide, heptad repeat 1 and 2, and a transmembrane domain, all of which are required to mediate fusion of the viral and host cell membranes.
[0007] SARS-CoV and SARS-CoV-2 bind to and use angiotensin-converting enzyme 2 (ACE2) of a host cell as a receptor to enter the host cells (Ge X.Y. et al, Nature. 2013 Nov 28;503(7477):535-8; Hoffmann M. et al, Cell. 2020 Mar 4. pii: S0092-8674(20)30229-4). The motif within the RBD that particularly binds to RCE2 is often referred to as the “ACE2-binding motif’. SARS-CoV can also use CD209L (also known as L-SIGN) as an alternative receptor (Jeffers S. A. et al, Proc Natl Acad Sci U
S A. 2004 Nov 2;101(44):15748-53). In contrast, MERS-CoV binds dipeptidyl peptidase 4 (“DPP4”, also known as CD26) of the host cell via a different RBD of the S protein.
[0008] Cell entry of coronaviruses often depends also on priming of the S protein by host cell proteases. Recently, SARS-CoV-2 was found to use the serine protease TMPRSS2 for S protein priming and ACE2 for entry (Wu A. et al, Cell Host Microbe. 2020 Mar ll;27(3):325-328;
Hoffmann M. et al, Cell. 2020 Mar 4. pii: S0092-8674(20)30229-4).
[0009] The genome of SARS-CoV-2 is about 29.8 kb nucleotides and encodes 15 nsps, four structural proteins (S, E, M, and N) and eight accessory proteins (3a, 3b, p6, 7a, 7b, 8b, 9b, and orfl4) (Wu A. et al, Cell Host Microbe. 2020 Mar ll;27(3):325-328). While SARS-CoV-2 is genetically close to a SARS-like bat CoV and also to SARS-CoV, a number of sequence differences have been identified. When SARS-CoV-2 is compared to SARS-CoV or SARS-like bat CoV, 380 amino acid differences or substitutions were found, 27 of which are in the S protein, including 6 substitutions in the RBD at amino acid region 357-528 (but not in the receptor-binding motifs that directly interact with ACE2) and 6 substitutions in the underpinning subdomain (SD) at amino acid region 569-655. [0010] One of the few drugs approved by the U.S. Food and Drug Administration (“FDA”) for use in treating COVID-19 is the viral replication inhibitor remdesivir. Clinical trials demonstrated that remdesivir shortens the time to recovery in hospitalized patients, but more effective therapy is in great need. Convalescent plasma received the emergency use authorization status by the FDA. Other treatments given to COVID-19 patients include anti-inflammatories such as corticosteroids and other treatments for managing symptoms such as supplemental oxygen and mechanical ventilatory support. Several drugs, particularly those that have been approved for preventing or treating other infectious disease, are currently being tested in the clinic, which includes e.g., lopinavir-ritonavir (HIV protease inhibitor), ABX464 (viral RNA splicer), favilavir (RNA-dependent RNA polymerase inhibitor used for influenza virus infection), niclosamide and ivermectin (antihelmintic), and BCG vaccine (vaccine for tuberculosis). Also, other ongoing clinical trials reportedly are using IL-6 receptor antagonist antibodies, an anti-GM-CSF or anti-GM-CSF receptor antibody, an anti-TNF antibody, an anti-IL- lbeta antibody, or an anti-complement component 5 antibody, in an effort to inhibit inflammation and thereby potentially inhibit cytokine storm and sepsis which can manifest in some SARS-CoV-2- infected patients and may cause death.
SUMMARY
[0011] In one aspect, the present disclosure relates to a compound which binds to coronavirus (CoV) or the spike protein (S protein) of a CoV (“CoV-S”). In some embodiments, the compound may be an isolated antibody or antigen-binding antibody fragment which binds to a CoV-S. In some embodiments, the antibody or antigen-binding antibody fragment may comprise a heavy chain variable region (VH), or fragments thereof, and/or a light chain variable region (VL), or fragments
thereof. In certain embodiments, the VH or fragment thereof may comprise a complementarity determining region 1 (CDR1), a complementarity-determining region 2 (CDR2), and a complementarity-determining region 3 (CDR3), which may also be referred to as VH CDR1, VH CDR2, and VH CDR3, respectively. In certain embodiments, the VL or fragment thereof may comprise a CDR1, a CDR2, and a CDR3, which may also be referred to as VL CDR1, VL CDR2, and VL CDR3, respectively. In some embodiments, the antibody, or antigen-binding antibody fragment thereof, may comprise a heavy chain CDR1, a heavy chain CDR2, a heavy chain CDR3, a light chain CDR1, a light chain CDR2, and a light chain CDR3.
[0012] In some embodiments, the antibody or antigen-binding antibody fragment may comprise an antibody or antigen-binding antibody fragment thereof, or an affinity-matured variant of an anti-Co V- S antibody or antigen-binding antibody fragment thereof; selected from the group consisting of ADI- 58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI- 58128, ADI-58129, ADI-58130, ADI-58131, optionally wherein the CoV-S is SARS-CoV-S or SARS-CoV-2-S.
[0013] In some embodiments, the antibody, or antigen-binding antibody fragment thereof, may comprise a VH and/or VL. In certain embodiments, the VH may comprise a CDR3 having an amino acid sequence identical to the VH CDR3 of any one of anti-CoV-S antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, ADI-58131, and optionally, the VL CDR3 may comprise a CDR3 having an amino acid sequence identical to the VL CDR3 of the same anti-CoV-S antibody that the VH CDR3 is derived from, and the anti-CoV-S antibody may be selected from any one of anti-CoV-S antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI- 58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI- 58130, ADI-58131. Here, the CoV-S may be the spike protein (“S protein”) of Severe Acute Respiratory Syndrome (SARS) coronavirus (“SARS-CoV”), which may be referred to as “SARS- CoV-S”, or the S protein of SARS-CoV -2 (also known as “n2019-nCoV”), which may be referred to as “SARS-CoV -2-S”. Optionally, the CoV-S may comprise a sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% identity to, comprising, or consisting of the amino acid sequence of SEQ ID NO: 401 (SARS-CoV-S, 1288 amino acids, Accession# PDB: 6VSB_B) or having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% identity to, comprising, or consisting of SEQ ID NO: 403 (SARS-CoV -2-S, 1273 amino acids, GenBank: QHD43416.1).
[0014] In some embodiments, the SARS-CoV -2-S is a B.1.1.7 variant, a B. 1.351 variant, a B.1.1.28 variant, a B. 1.429 variant, a P.l varaint, a B.1.617 variant ( e.g ., B.1.617.1 and B.1.617.2), a C.37 variant, a 1.621 variant, a AY.l variant, a 1.623 variant, a C.36 variant, a A.27 variant, a AV.l variant, a B.1.1.482 variant, a B.1.1.523 variant, a B.1.427 variant, a AY.4 variant, a AY.ll variant,
variant, a D614G variant of SEQ ID NO: 403, a B.1.1.529/BA.1 variant (also known as the Omicron variant), a BA.1.1 variant, or a BA.2 variant.
[0015] In some embodiments, the antibody or antigen-binding antibody fragment, optionally an affinity-matured variant of any of the anti-CoV-S antibodies disclosed herein, may comprise at least 1, 2, 3, 4, 5 or all 6 complementarity-determining regions (CDRs) of any one of anti-CoV-S antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, ADI-58131, optionally wherein the CoV-S is SARS-CoV-S or SARS-CoV-2-S. Optionally, the CoV-S may comprise the amino acid sequence of SEQ ID NO: 401 (SARS-CoV-S, 1288 amino acids, Accession# PDB: 6VSB_B) or SEQ ID NO: 403 (SARS-CoV-2-S, 1273 amino acids, GenBank: QHD43416.1). [0016] In some embodiments, the isolated antibody or antigen-binding antibody fragment, optionally an affinity-matured variant of any of the anti-CoV-S antibodies disclosed herein, may comprise: (a) a VH CDR1 polypeptide; (b) a VH CDR2 polypeptide; (c) a VH CDR3 polypeptide; (d) a VL CDR1 polypeptide; (e) a VL CDR2 polypeptide; and (f) a VL CDR3 polypeptide. The amino acid sequences of the VH CDR1, the VH CDR2, the VH CDR3, the VL CDR1, the VL CDR2, and the VL CDR3 may be identical to the amino acid sequences of the VH CDR1, VH CDR2, VH CDR3, VL CDR1,
VL CDR2, and VL CDR3, respectively, of any one of anti-CoV-S antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, ADI-58131. Optionally, the CoV-S may be SARS- CoV-S or of “SARS-CoV-2-S”. Lurther optionally, the CoV-S may comprise the amino acid sequence of SEQ ID NO: 401 (SARS-CoV-S, 1288 amino acids, Accession# PDB: 6VSB_B) or SEQ ID NO: 403 (SARS-CoV-2-S, 1273 amino acids, GenBank: QHD43416.1).
[0017] In certain embodiments, the isolated antibody or antigen-binding antibody fragment, optionally an affinity-matured variant of any of the anti-CoV-S antibodies disclosed herein, which specifically binds to CoV-S, may comprise: (a) a VH comprising a VH CDR1, VH CDR2, and VH CDR3; and (b) a VL comprising a VL CDR1, VL CDR2, and VL CDR3.
[0018] In some exemplary embodiments, the amino acid sequences of the VH CDR1, the VH CDR2, the VH CDR3, the VL CDR1, the VL CDR2, and the VL CDR3 may be identical to the amino acid sequences of: (1) SEQ ID NOS: 2, 4, 6, 202, 204, and 206, respectively; (2) SEQ ID NOS: 12, 14, 16, 212, 214, and 216, respectively; (3) SEQ ID NOS: 22, 24, 26, 222, 224, and 226, respectively; (4)
SEQ ID NOS: 32, 34, 36, 232, 234, and 236, respectively; (5) SEQ ID NOS: 42, 44, 46, 242, 244, and 246, respectively; (6) SEQ ID NOS: 52, 54, 56, 252, 254, and 256, respectively; (7) SEQ ID NOS: 62, 64, 66, 262, 264, and 66, respectively; (8) SEQ ID NOS: 72, 74, 76, 272, 274, and 276, respectively; (9) SEQ ID NOS: 82, 84, 86, 282, 284, and 286, respectively; (10) SEQ ID NOS: 92, 94, 96, 292,
294, and 296, respectively; (11) SEQ ID NOS: 102, 104, 106, 302, 304, and 306, respectively; or (12) SEQ ID NOS: 112, 114, 116, 312, 314, and 316, respectively.
[0019] In other words, the amino acid sequences of the VH CDR1, the VH CDR2, the VH CDR3, the VL CDR1, the VL CDR2, and the VL CDR3 may be identical to the VH CDR1, the VH CDR2, the VH CDR3, the VL CDR1, the VL CDR2, and the VL CDR3 and VL amino acid sequences of any one of anti-CoV-S antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, ADI-58131.
[0020] In some embodiments, the isolated antibody or antigen-binding antibody fragment, optionally an affinity-matured variant of any of the anti-CoV-S antibodies disclosed herein, may possess one of the following structural features:
(1) (a) the VH may comprise an amino acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 8, and (b) the VL may comprise an amino acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 208;
(2) (a) the VH may comprise an amino acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 18, and (b) the VL may comprise an amino acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 218;
(3) (a) the VH may comprise an amino acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 28, and (b) the VL may comprise an amino acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 228;
(4) (a) the VH may comprise an amino acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 38, and (b) the VL may comprise an amino acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 238;
(5) (a) the VH may comprise an amino acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 48, and (b) the VL may comprise an amino acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 248;
(6) (a) the VH may comprise an amino acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 58, and (b) the VL may comprise an amino acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 258;
(7) (a) the VH may comprise an amino acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 68, and (b) the VL may
comprise an amino acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 268;
(8) (a) the VH may comprise an amino acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 78, and (b) the VL may comprise an amino acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 278;
(9) (a) the VH may comprise an amino acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 88, and (b) the VL may comprise an amino acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 288;
(10) (a) the VH may comprise an amino acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 98, and (b) the VL may comprise an amino acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 298;
(11) (a) the VH may comprise an amino acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 108, and (b) the VL may comprise an amino acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 308; or
(12) (a) the VH may comprise an amino acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 118, and (b) the VL may comprise an amino acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 318.
[0021] In some exemplary embodiments, the isolated antibody or antigen-binding antibody fragment, optionally an affinity-matured variant, may be human, humanized, primatized or chimeric.
[0022] In some exemplary embodiments, the isolated antibody or antigen-binding antibody fragment, optionally an affinity-matured variant, may be bispecific or multispecific.
[0023] In some exemplary embodiments, the isolated antibody or antigen-binding antibody fragment, optionally an affinity-matured variant, may comprise at least one first antigen-binding domain (“ABD”) and at least one second ABD.
[0024] Here, the following features (a) and (b) may be met:
(a) the first ABD may comprise the VH CDR1, the VH CDR2, the VH CDR3, the VL CDR1, the VL CDR2, and the VL CDR3 of a first anti-CoV-S antibody selected from any one of anti-CoV-S antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, ADI-58131; and/or
(b) the second ABD may comprise the VH CDR1, the VH CDR2, the VH CDR3, the VL CDR1, the VL CDR2, and the VL CDR3 of a second anti-CoV-S antibody selected from any one of anti-CoV-S
antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, ADI-58131. [0025] Optionally, the first anti-CoV-S antibody may be same as the second anti-CoV-S antibody or may be different from the second anti-CoV-S antibody.
[0026] The first anti-CoV-S antibody and the second anti-CoV-S antibody may bind to the same or different coronavirus species. Optionally, the first CoV-S and the second CoV-S may be (i) both of SARS-CoV or (ii) both of SARS-CoV-2.
[0027] Further optionally, the first anti-CoV-S antibody may be same as the second anti-CoV-S antibody or may be different from the second anti-CoV-S antibody. Still further optionally, these antibodies may bind to the same or different epitopes on a CoV-S expressed by said SARS-CoV or SARS-CoV-2. Alternatively, the first anti-CoV-S antibody and the second anti-CoV-S antibody may bind to different coronaviruses, optionally wherein the first CoV-S and the second CoV-S are (i) SARS-CoV and of SARS-CoV-2 coronaviruses, respectively, or are (ii) SARS-CoV-2 and of SARS- CoV coronaviruses, respectively.
[0028] In some embodiments, the bispecific or multispecific isolated antibody or antigen-binding antibody fragment may comprise at least one first ABD and at least one second ABD.
[0029] In certain embodiments, (a) the first ABD may comprise the VH CDR1, the VH CDR2, the VH CDR3, the VL CDR1, the VL CDR2, and the VL CDR3 of a first anti-CoV-S antibody selected from any one of anti-CoV-S antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, ADI-58131, or an affinity-matured variant of any of the foregoing; and/or (b) the second ABD binds to an antigen which may not be a CoV-S, optionally wherein the antigen is a cytokine, a cytokine receptor, or an immunomodulatory polypeptide.
[0030] In some embodiments, the isolated antibody or antigen-binding antibody fragment may comprise a Fab, Fab’, F(ab’)2, scFv, sc(Fv)2, minibody, diabody, sdAb, BITE.
[0031] In some embodiments, the isolated antibody or antigen-binding antibody fragment may comprise a constant region or Fc region or at least one domain thereof.
[0032] In certain embodiments, the constant region or Fc region may comprise a mutation which impairs or enhances at least one effector function, optionally FcR binding, FcRn binding, complement binding, glycosylation, complement-dependent cytotoxicity (“CDC”), or antibody-dependent cellular cytotoxicity (“ADCC”).
[0033] In some embodiments, the constant or Fc region is primate derived, preferably human.
[0034] The human constant or Fc region optionally may be selected from a human IgGl, IgG2, IgG3 or IgG4 constant or Fc region which optionally may be modified, optionally such as by domain deletion or by introducing one or more mutations which impair or enhance at least one effector function.
[0035] The present disclosure further relates to chimeric antigen receptors (“CARs”) comprising at least one antibody or antigen-binding antibody fragment described herein.
[0036] The present disclosure further relates to antibody-drug conjugates (“ADCs”) comprising: (a) at least one antibody or antigen-binding antibody fragment described herein; and (b) a drug.
[0037] In some embodiments, the drug may be: (i) an antiviral drug, which is optionally, remdesivir, favipiravir, darunavir, nelfinavir, saquinavir, lopinavir or ritonavir; (ii) an antihelminth drug, which may be optionally ivermectin; (iii) an antiparasite drug, which may be optionally hydroxychloroquine, chloroquine, or atovaquone; (iv) antibacterial vaccine, which may be optionally the tuberculosis vaccine BCG; or (v) an anti-inflammatory drug, which may be optionally a steroid such as ciclesonide, a TNF inhibitor (e.g., adalimumab), a TNF receptor inhibitor (e.g., etanercept), an IL-6 inhibitor (e.g., clazakizumab), an IL-6 receptor inhibitor (e.g., toclizumab), or metamizole; (vi) an antihistamine drug, which may be optionally bepotastine; (vii) an ACE inhibitor, which may be optionally moexipril; (viii) a drug that inhibits priming of CoV-S, which may be optionally a serine protease inhibitor such as nafamostat; or (ix) a cytotoxic drug, which may be optionally daunorubicin, mitoxantrone, doxorubicin, cucurbitacin, chaetocin, chaetoglobosin, chlamydocin, calicheamicin, nemorubicin, cryptophyscin, mensacarcin, ansamitocin, mitomycin C, geldanamycin, mechercharmycin, rebeccamycin, safracin, okilactomycin, oligomycin, actinomycin, sandramycin, hypothemycin, polyketomycin, hydroxyellipticine, thiocolchicine, methotrexate, triptolide, taltobulin, lactacystin, dolastatin, auristatin, monomethyl auristatin E (MMAE), monomethyl auristatin F (MMAF), telomestatin, tubastatin A, combretastatin, maytansinoid, MMAD, MMAF, DM1, DM4, DTT, 16-GMB -APA-GA, 17-DMAP-GA, JW 55, pyrrolobenzodiazepine, SN-38, Ro 5-3335, puwainaphycin, duocarmycin, bafilomycin, taxoid, tubulysin, ferulenol, lusiol A, fumagillin, hygrolidin, glucopiericidin, amanitin, ansatrienin, cinerubin, phallacidin, phalloidin, phytosphongosine, piericidin, poronetin, phodophyllotoxin, gramicidin A, sanguinarine, sinefungin, herboxidiene, microcolin B, microcystin, muscotoxin A, tolytoxin, tripolin A, myoseverin, mytoxin B, nocuolin A, psuedolaric acid B, pseurotin A, cyclopamine, curvulin, colchicine, aphidicolin, englerin, cordycepin, apoptolidin, epothilone A, limaquinone, isatropolone, isofistularin, quinaldopeptin, ixabepilone, aeroplysinin, arruginosin, agrochelin, or epothilone.
[0038] The present disclosure also relates to isolated nucleic acids encoding any of the antibodies or antigen-binding antibody fragments disclosed herein.
[0039] In some embodiments, the nucleic acid may comprise:
(1) (a) a nucleic acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the nucleic acid sequence of SEQ ID NO: 9, and/or (b) a nucleic acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the nucleic acid sequence of SEQ ID NO: 209;
(2) (a) a nucleic acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the nucleic acid sequence of SEQ ID NO: 19, and/or (b) a nucleic acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the nucleic acid sequence of SEQ ID NO: 219;
(3) (a) a nucleic acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the nucleic acid sequence of SEQ ID NO: 29, and/or (b) a nucleic acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the nucleic acid sequence of SEQ ID NO: 229;
(4) (a) a nucleic acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the nucleic acid sequence of SEQ ID NO: 39, and/or (b) a nucleic acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the nucleic acid sequence of SEQ ID NO: 239;
(5) (a) a nucleic acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the nucleic acid sequence of SEQ ID NO: 49, and/or (b) a nucleic acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the nucleic acid sequence of SEQ ID NO: 249;
(6) (a) a nucleic acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the nucleic acid sequence of SEQ ID NO: 59, and/or (b) a nucleic acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the nucleic acid sequence of SEQ ID NO: 259;
(7) (a) a nucleic acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the nucleic acid sequence of SEQ ID NO: 69, and/or (b) a nucleic acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the nucleic acid sequence of SEQ ID NO: 269;
(8) (a) a nucleic acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the nucleic acid sequence of SEQ ID NO: 79, and/or (b) a nucleic acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the nucleic acid sequence of SEQ ID NO: 279;
(9) (a) a nucleic acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the nucleic acid sequence of SEQ ID NO: 89, and/or (b) a nucleic acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the nucleic acid sequence of SEQ ID NO: 289;
(10) (a) a nucleic acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the nucleic acid sequence of SEQ ID NO: 99, and/or (b) a nucleic acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the nucleic acid sequence of SEQ ID NO: 299;
(11) (a) a nucleic acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the nucleic acid sequence of SEQ ID NO: 109, and/or (b) a nucleic acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the nucleic acid sequence of SEQ ID NO: 309; or
(12) (a) a nucleic acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the nucleic acid sequence of SEQ ID NO: 119, and/or (b) a nucleic acid sequence with at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the nucleic acid sequence of SEQ ID NO: 319.
[0040] The present disclosure also relates to isolated cells which may comprise any of the nucleic acids disclosed herein.
[0041] In some embodiments the cell may be a bacterial, yeast, insect, fungal, or mammalian cell, optionally a human cell, further optionally a CHO or HEK cell.
[0042] In some embodiments the cell may be a human immune cell, optionally a T, NK, B or dendritic cell.
[0043] The present disclosure further relates to methods of expressing the antibody or antigen binding antibody fragment or the CAR disclosed herein.
[0044] In some embodiments, the method may comprise: (a) culturing the cell expressing an antibody or antigen-binding antibody fragment or CAR of the present disclosure under conditions that permit expression; and (b) optionally isolating the antibody or antigen-binding antibody fragment or the CAR from the cell or the culture medium containing the cell.
[0045] The present disclosure further relates to methods of identifying an antibody or an antigen binding antibody fragment which specifically binds to CoV-S.
[0046] In some embodiments, the method may comprise: (a) obtaining antisera and/or B cells obtained from a patient infected with SARS-CoV or SARS-CoV-2, optionally wherein the patient recovered from SARS-CoV or SARS-CoV-2 infection or the patient is a convalescent patient infected with SARS-CoV or SARS-CoV-2; (b) contacting the antisera and/or B cells with the CoV-S; and (c) isolating an antibody or antigen-binding fragment thereof which specifically bind to the CoV-S. Optionally, the CoV-S is the spike protein of SARS-CoV (“SARS-CoV-S”) or of SARS-CoV-2 (“SARS-CoV-2-S”). Further optionally, the CoV-S may comprise the amino acid sequence of SEQ ID NO: 401 (SARS-CoV-S, 1288 amino acids, Accession# PDB: 6VSB_B) or SEQ ID NO: 403 (SARS- CoV -2-S, 1273 amino acids, GenBank: QHD43416.1).
[0047] In some embodiments, the method may further detect that the antibody or antigen-binding fragment thereof which specifically binds to CoV-S neutralizes, blocks or inhibits coronavirus infectivity or coronavirus proliferation, optionally wherein the coronavirus is SARS-CoV or SARS- CoV-2.
[0048] In certain embodiments, the method may further detect whether the antibody or antigen binding antibody fragment thereof which specifically binds to the CoV-S binds to other coronaviruses, optionally selected from the group consisting of MERS-CoV, HCoV-HKUl, HCoV- OC43, HCoV-229E, and HCoV-NL63.
[0049] In any of such detection methods, the method may further comprise determining the sequence of the antibody or antigen-binding antibody fragment thereof may be determined.
[0050] In some embodiments these sequences may be affinity-matured or mutated to enhance binding affinity and/or potentially increase specificity to a particular CoV-S.
[0051] The present disclosure further provides compositions comprising: (a) at least one antibody or antigen-binding antibody fragment of the present disclosure; and (b) a pharmaceutically acceptable carrier or excipient.
[0052] The present disclosure further provides methods of determining whether a subject has been infected with SARS-CoV or SARS-CoV-2 or another coronavirus by detecting whether a biological sample from the subject may comprise SARS-CoV-S protein or SARS-CoV-S-2 protein or another coronavirus S protein homologous thereto based on its immunoreaction with at least one antibody or antigen-binding antibody fragment disclosed herein. The sample may optionally be blood, plasma, lymph, mucus, urine, and/or feces. Optionally, the SARS-CoV S may comprise the amino acid sequence of SEQ ID NO: 401 (SARS-CoV-S, 1288 amino acids, Accession# PDB: 6VSB_B),
[0053] Alternatively, the SARS-CoV-2 may comprise the amino acid sequence of SEQ ID NO: 403 (SARS-CoV -2-S, 1273 amino acids, GenBank: QHD43416.1).
[0054] Such determination methods optionally may comprise an ELISA or radioimmunoassay.
[0055] In such determination methods, the subject optionally may be human, a companion animal (e.g., a dog or cat), an agricultural animal, e.g., animals used in meat production, or may comprise an animal in a zoo, e.g. , a tiger or lion.
[0056] In such determination methods, the samples optionally may be collected at different times from the subject and the presence or absence or the level of SARS-CoV-S or SARS-CoV-S-2 or another coronavirus S protein homologous thereto may be detected in order to assess whether the subject has recovered. Here, the SARS-CoV S may comprise the amino acid sequence of SEQ ID NO: 401 (SARS-CoV-S, 1288 amino acids, Accession# PDB: 6VSB_B), and optionally the SARS-CoV-2 may comprise the amino acid sequence of SEQ ID NO: 403 (SARS-CoV -2-S, 1273 amino acids, GenBank: QHD43416.1).
[0057] The present disclosure further provides methods of inducing an immune response against SARS-CoV or SARS-CoV-2 or another coronavirus, which may be selected from MERS-CoV, HCoV-HKUl, HCoV-OC43, HCoV-229E, and HCoV-NL63, in a subject in need thereof.
[0058] In some embodiments, the methods may comprise administering at least one antibody or antigen-binding antibody fragment of the present disclosure.
[0059] In some embodiments, the methods may comprise administering a cocktail of different antibodies or antigen-binding antibody fragments of the present disclosure, e.g., which bind to the same or different epitopes on the same or different CoV-Ss.
[0060] In certain embodiments, the immune response elicits immunoprotection, optionally prolonged, against at least one coronavirus, optionally SARS-CoV or SARS-CoV-2, further optionally against another coronavirus.
[0061] The present disclosure further provides methods of inhibiting or blocking infection of susceptible cells by SARS-CoV or SARS-CoV-2 or another coronavirus, such as MERS-CoV, HCoV- HKU1, HCoV-OC43, HCoV-229E, and HCoV-NL63, in a subject in need thereof.
[0062] In some embodiments, the method may comprise administering at least one antibody or antigen-binding antibody fragment, of the present disclosure, e.g., a cocktail as above-described. [0063] The present disclosure further provides methods of treating infection by SARS-CoV or SARS-CoV-2 or another coronavirus optionally such as MERS-CoV, HCoV-HKUl, HCoV-OC43, HCoV-229E, and HCoV-NL63, or treating a condition, symptom, disease, or disorder associated with said infection in a subject in need thereof.
[0064] In some embodiments, the method may comprise administering to the subject a therapeutically effective amount of at least one antibody or antigen-binding antibody fragment, an ADC or a CAR, of the present disclosure, e.g., a cocktail as above-described.
[0065] In some embodiments, the condition, symptom, disease, or disorder comprises at least one of bronchitis, pneumonia, respiratory failure, acute respiratory failure, organ failure, multi-organ system failure, pediatric inflammatory multisystem syndrome, acute respiratory distress syndrome, blood clot, a cardiac condition, myocardial injury, myocarditis, heart failure, cardiac arrest, acute myocardial infarction, dysrhythmia, venous thromboembolism, post-intensive care syndrome, shock, anaphylactic shock, cytokine release syndrome, septic shock, disseminated intravascular coagulation, ischemic stroke, intracerebral hemorrhage, microangiopathic thrombosis, psychosis, seizure, nonconvulsive status epilepticus, traumatic brain injury, stroke, anoxic brain injury, encephalitis, posterior reversible leukoencephalopathy, necrotizing encephalopathy, post-infectious encephalitis, autoimmune mediated encephalitis, acute disseminated encephalomyelitis, acute kidney injury, acute liver injury, pancreatic injury, immune thrombocytopenia, subacute thyroiditis, a gastrointestinal complication, aspergillosis, increased susceptibility to infection with another virus or bacteria, and/or a pregnancy-related complication.
[0066] The present disclosure also provides methods of preventing infection by SARS-CoV or SARS-CoV-2 or another coronavirus optionally selected from the group consisting of MERS-CoV, HCoV-HKUl, HCoV-OC43, HCoV-229E, and HCoV-NL63 in a subject in need thereof.
[0067] In some embodiments, the method may comprise administering to the subject a prophylactically effective amount of at least one antibody or antigen-binding antibody fragment, an ADC or a CAR, of the present disclosure, e.g., a cocktail as above-described.
[0068] The present disclosure also provides methods of preventing the need for a subject infected with SARS-CoV or SARS-CoV-2 or another coronavirus optionally selected from the group consisting of MERS-CoV, HCoV-HKUl, HCoV-OC43, HCoV-229E, and HCoV-NL63 to be placed on a ventilator, or reducing the time that a subject infected with SARS-CoV or SARS-CoV-2 or another coronavirus optionally selected from the group consisting of MERS-CoV, HCoV-HKUl, HCoV-OC43, HCoV-229E, and HCoV-NL63 is on a ventilator.
[0069] In some embodiments, the method may comprise administering to the subject a prophylactically or therapeutically effective amount of at least one antibody or antigen-binding antibody fragment, an ADC or a CAR, of the present disclosure, e.g., a cocktail as above-described. [0070] The present disclosure provides methods of preventing the onset of pneumonia in a subject infected SARS-CoV or SARS-CoV-2 or another coronavirus optionally selected from the group consisting of MERS-CoV, HCoV-HKUl, HCoV-OC43, HCoV-229E, and HCoV-NL63, or treating pneumonia and/or the symptoms of pneumonia in a subject for a subject infected SARS-CoV or SARS-CoV-2 or another coronavirus optionally selected from the group consisting of MERS-CoV, HCoV-HKUl, HCoV-OC43, HCoV-229E, and HCoV-NL63.
[0071] In some embodiments, the antibody, or antigen-binding fragment thereof, is administered within 5 days of symptom onset. In some embodiments, the antibody, or antigen-binding fragment thereof, is administered within 4 days of symptom onset. In some embodiments, the antibody, or antigen-binding fragment thereof, is administered within 3 days of symptom onset. In some embodiments, the antibody, or antigen-binding fragment thereof, is administered within 2 days of symptom onset. In some embodiments, the antibody, or antigen-binding fragment thereof, is administered within 1 day of symptom onset.
[0072] In some embodiments, the method may comprise administering to the subject a prophylactically or therapeutically effective amount of at least one antibody or antigen-binding antibody fragment, an ADC or a CAR, of the present disclosure, e.g., a cocktail as above-described. [0073] In any of such methods, the subject optionally may be human or may comprise a companion animal, agricultural animal or animal in a zoo.
[0074] In some embodiments, the subject is an adult. In some embodiment, the subject is an adolescent. In some embodiments, the subject is a child, or a pediatric subject. In one embodiment, the subject is a child, e.g., from birth to age of 11 years old, e.g., birth to < 1 month, 1 month to <2 years, 5-11 years old, or 2-11 years old. In other embodiments, the adolescent is 12-17 years old or 12-15 years old. In one embodiment, the adult is over the age of 18. In some embodiments, the subject has no known recent exposure to COVID-19. In some embodiments, the subject has a known
recent exposure to COVID-19. In one embodiment, recent exposure is within the last 14 days, within the last 10 days, within the last 7 days, within the last 6 days, 5 days, 4 days, 3 days, 2 days, or 24 hours. In some embodiments, the subject has COVID-19 symptoms or an active COVID-19 infection. In some embodiments, the subject, e.g., the adult, the adolescent, or the child, is vaccinated or has received a COVID-19 vacination. In some embodiments, the subject, e.g., the adult, the adolescent, or the child, is not vaccinated or has not received a COVID-19 vacination.
[0075] In some embodiments, the subject is immunocompromised. In some embodiments, the subject is a significantly immune compromised subject, e.g., adult subject or pediatric subject, but whose underlying medical condition(s) put them at increased risk of developing severe symptomatic COVID-19. In some embodiments, the subject has no known recent exposure to COVID-19. In some embodiments, the subject has a known recent exposure to COVID-19. In one embodiment, recent exposure is within the last 14 days, within the last 10 days, within the last 7 days, within the last 6 days, 5 days, 4 days, 3 days, 2 days, or 24 hours. In some embodiments, the subject has COVID-19 symptoms or an active COVID-19 infection. In some embodiments, the immunocompromsed subject includes, but is not limited to, an individual who is actively treated for solid tumor or hematologic malignancies; solid organ transplant (SOT) recipient taking immunosuppressive therapy; CAR-T-cell therapy or HCT recipient; subjects having moderate or severe primary immunodeficiency; subjects having advanced or untreated HIV infection, or who are taking high-dose corticosteroids, alkylating agents, antimetabolites, or TNF blockers. In some embodiments, the immunocompromised subject is vaccinated or has received a COVID-19 vacination. In some embodiments, the immunocompromised subject is not vaccinated or has not received a COVID-19 vacination.
[0076] In some embodiments, the subject is at a high risk of COVID progression. In some embodiments, the subjects at a high risk of disease progression are age 50 years old or above with no comorbid conditions or additional risk factors for progression of COVID-19.
[0077] In some embodiments, the subjects at a high risk of disease progression are those about 18 to about 50 years old and with one or more preexisting medical conditons selected from the group consisting of (i) obesity (body mass index (BMI) > 30 kg/m2), (ii) diabetes (type 1 or type 2); (iii) chronic kidney disease (eGFR calculated by modification of diet in renal disease (MDRD) of 59 mL/min/1.73 m2 or less, including end-stage renal disease on hemodialysis); (iv) chronic lung disease (emphysema/chronic obstructive pulmonary disease, chronic bronchitis, interstitial lung disease [including idiopathic pulmonary fibrosis], cystic fibrosis, or moderate to severe asthma [defined as requiring daily therapy]); (v) cardiac disease (heart failure, coronary artery disease, cardiomyopathies, or hypertension [with at least one medication prescribed or recommended]); (vi) sickle cell disease or thalassemia; (vii) solid organ or blood stem cell transplant recipients; (viii) other immunodeficiency due to underlying illness or immunosuppressant medication (e.g., corticosteroids >20 mg/day
prednisone or equivalent); (ix) Down Syndrome, (x) stroke or cerebrovascular disease, which affects blood flow to the brain; (xi) substance use disorder; or (xii) pregnant.
[0078] In some embodiments, the subjects at a high risk of disease progression are those about 12 to about 17 years old (inclusive) and with one or more preexisting medical conditions selected from the group consisting of (i) BMI >85th percentile for age and sex based on United States Center for Disease Control (CDC) growth charts; (ii) diabetes (Type 1 or Type 2); (iii) chronic kidney disease; (iv) sickle cell disease or thalassemia; (v) congenital or acquired heart disease; (vi) neurodevelopmental disorders (e.g., cerebral palsy, Down syndrome); (vii) a medically-related technological dependence (e.g., tracheostomy, gastrostomy, or positive pressure ventilation not related to COVID-19); (viii) asthma, reactive airway or other chronic respiratory disease that requires daily medication for control; (ix) solid organ or blood stem cell transplant recipients; (x) other immunodeficiency due to underlying illness or immunosuppressant medication; (xi) substance use disorder; or (xii) pregnant.
[0079] In some embodiments, the subject is age 50 or above with no comorbid conditions or additional risk factors for progression of COVID-19.
[0080] In some embodiments, the subject has hypertension with at least one medication prescribed or recommended.
[0081] In some embodiments, the subject has moderate to severe asthma requiring daily therapy. [0082] Optionally the subject may have at least one risk factor which renders them more prone to a poor clinical outcome.
[0083] In certain embodiments, wherein the risk factors may comprise one or more of (i) advanced age such as over 55, 60 or 65 years old, (ii) diabetes, (iii) a chronic respiratory condition such as asthma, cystic fibrosis, another fibrotic condition, or COPD, (iv) obesity, (iv) hypertension, (v) a cardiac or cardiovascular condition, such as heart defects or abnormalities, (vi) a chronic inflammatory or autoimmune condition, e.g., lupus or multiple sclerosis, and (vii) an immunocompromised status which optionally may be caused by cancer, chemotherapy, smoking, bone marrow or organ transplantation, immune deficiencies, poorly controlled HIV infection or AIDS, or prolonged use of corticosteroids or other immunosuppressive medications.
In certain embodiments, the subject may further be treated with at least one other drug. In certain embodiments, the method further comprises administering to the subject at least one other drug. Optionally, such one other drug may be: (i) an antiviral drug, optionally, remdesivir, favipiravir, darunavir, nelfinavir, saquinavir, lopinavir, or ritonavir; (ii) an antihelminth drug, optionally ivermectin; (iii) an antiparasitic drug, optionally hydroxychloroquine, chloroquine, or atovaquone;
(iv) antibacterial vaccine, optionally the tuberculosis vaccine BCG; (v) an anti-inflammatory drug, optionally a steroid such as ciclesonide, a TNF inhibitor (e.g., adalimumab), a TNF receptor inhibitor (e.g., etanercept), an IL-6 inhibitor (e.g., clazakizumab), an IL-6 receptor inhibitor (e.g., tocilizumab),
or metamizole; (vi) an antihistamine drug, optionally bepotastine; (vii) an ACE inhibitor, optionally moexipril; and/or (viii) a drug that inhibits priming of CoV-S, optionally a serine protease inhibitor, further optionally nafamostat.
[0084] In certain embodiments, the subject may further be treated with: (I) an antiviral agent, optionally, remdesivir, favipiravir, darunavir, nelfinavir, saquinavir, lopinavir, or ritonavir; and (II) at least one other drug. In certain embodiments, the method may further comprise administering to the subject (I) an antiviral agent, optionally, remdesivir, favipiravir, darunavir, nelfinavir, saquinavir, lopinavir, or ritonavir; and (II) at least one other drug. Optionally, the at least one other drug may be (i) an antihelminth drug, further optionally ivermectin; (ii) an antiparasitic drug, optionally hydroxychloroquine, chloroquine, or atovaquone; (iii) an antibacterial vaccine, which is optionally the tuberculosis vaccine BCG; or (iv) an anti-inflammatory drug, optionally a steroid such as ciclesonide, a TNF inhibitor (e.g., adalimumab), a TNF receptor inhibitor (e.g., etanercept), an IF-6 inhibitor (e.g., clazakizumab), an IF-6 receptor inhibitor (e.g., toclizumab), or metamizole; (v) an antihistamine drug, optionally bepotastine; (vi) an ACE inhibitor, optionally moexipril; and/or (vii) a drug that inhibits priming of CoV-S, which is optionally a serine protease inhibitor such as nafamostat. In some embodiments, the subject may further be treated with a vaccine, or has been treated with a vaccine, e.g., a COVID-19 vaccine. In some embodiments, the anti-CoV-S antibodies or antigen-binding fragments thereof are administered as a vaccine supplement. As used herein, the term “vaccine supplement” refers to a treatment that is administered prior to, concurrently with, or after, receving a vaccine treatment, e.g., a COVID-19 vaccine. The vaccine supplement may be administered instead of a vaccine booster or an additional dose.
[0085] In some embodiments, the anti-CoV-S antibody, or antigen-binding fragment thereof, is administered in combination with a vaccine. In some embodiments, the anti-CoV-S antibody, or antigen-binding fragment thereof, is administered concurrently with a COVID-19 vaccine. In some embodiments, the anti-CoV-S antibody, or antigen-binding fragment thereof, is administered after a COVID-19 vaccine. In some embodiments, the anti-CoV-S antibody, or antigen-binding fragment thereof, is administered prior to COVID-19 vaccine.
[0086] In one embodiment, the anti-CoV-S antibodies, or antigen -binding fragments thereof, are administered concurrently with a COVID-19 vaccine, within 24 hours of a COVID-19 vaccine, within 48 hours of a COVID-19 vaccine, within 3 days, 4 days, 5 days, 6 days, 7 days, 10 days, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months or 12 months of a COVID-19 vaccine. In one embodiment, the COVID-19 vaccine is an mRNA vaccine. In one embodiment, the COVID- 19 vaccine is COMIRNATY (by PFIZER, also known as BNT 162b2), the Moderna COVID-19 vaccine (mRNA-1273), the Johnson & Johnson COVID-19 vaccine (JNJ-78436735), or the AstraZeneca COVID-19 vaccine.
[0087] In some embodiments, anti-CoV-S antibodies or antigen-binding antibody fragments of the present disclosure may be characterized by having a certain VH CDR3 sequences or having a VH CDR3 sequences that are similar to a certain VH CDR3.
[0088] In certain embodiments, antibody or antigen-binding antibody fragment of the present disclosure may comprise an Fc region. The Fc region may comprise a wild type sequence or a variant sequence and optionally may comprise an amino acid sequence of SEQ ID NOs: 411, 412, 413, 414, 415, 416, or 417.
[0089] In certain embodiments, the isolated antibody or antigen-binding antibody fragment may bind to the SI subunit of SARS-CoV-S or of SARS-CoV-2-S.
[0090] In certain embodiments, the isolated antibody or antigen-binding antibody fragment may bind to the receptor binding domain (RBD) or the N-terminal domain (NTD) of SARS-CoV-S or of SARS- CoV-2-S.
[0091] In certain embodiments, the isolated antibody or antigen-binding antibody fragment may bind to the ACE2-binding motif of SARS-CoV-S or of SARS-CoV-2-S and optionally further binds to the epitope of the antibody CR3022.
[0092] In further embodiments, the isolated antibody or antigen-binding antibody fragment may compete with ACE2.
[0093] In further embodiments, the isolated antibody or antigen-binding antibody fragment may compete with: (i) ACE2 and the antibody CR3022; or (ii) ACE2 but not the antibody CR3022. In certain embodiments, the isolated antibody or antigen-binding antibody fragment (a) may bind to the S protein of SARS-CoV and/or of SARS-CoV-2; and (b) may not bind to any of the S proteins of HCoV-229E, HCoV-HKUl, HCoV-NL63, and HCoV-OK43.
[0094] In certain embodiments, the isolated antibody or antigen-binding antibody fragment may (a) bind to the S protein of SARS-CoV and/or of SARS-CoV-2; and also (b) bind to the S protein of at least one of HCoV-229E, HCoV-HKUl, HCoV-NL63, and HCoV-OK43.
[0095] In further embodiments, the isolated antibody or antigen-binding antibody fragment may neutralize SARS-CoV and/or SARS-CoV-2.
[0096] In further embodiments, the isolated antibody or antigen-binding antibody fragment may neutralize SARS-CoV and/or SARS-CoV-2 at 100 nM in vitro.
[0097] In further embodiments, the isolated antibody or antigen-binding antibody fragment may neutralize SARS-CoV and/or SARS-CoV-2 at: (i) an IC50 of about 100 nM or lower, of about 50 nM or lower, of about 20 nM or lower, of about 10 nM or lower, of about 5 nM or lower, of about 2 nM or lower, of about 1 nM or lower, of about 500 pM or lower, of about 200 pM or lower, of about 100 pM or lower, of about 50 pM or lower, of about 20 pM or lower, of about 10 pM or lower, of about 5 pM or lower, of about 2 pM or lower, or of about 1 pM or lower; and/or (ii) an IC50 of about 5 μg/mL or lower, of about 4.5 μg/mL or lower, of about 4 μg/mL or lower, of about 3.5 μg/mL or
lower, of about 3 mg/mL or lower, of about 2.5 mg/mL or lower, about 2 mg/mL or lower, of about 1.5 mg/mL or lower, of about 1 mg/mL or lower, of about about 500 ng/mL or lower, of about 400 ng/mL or lower, of about 200 ng/mL or lower, of about 100 ng/mL or lower, of about 50 ng/mL or lower, of about 20 ng/mL or lower, of about 10 ng/mL or lower, of about 5 ng/mL or lower, of about 2 ng/mL or lower, or of about 1 ng/mL or lower, or about 0.6 ng/mL or lower, or of about 0.1 ng/mL to about 5 mg/mL, of about 0.5 ng/mL to about 4.5 mg/mL, of about 0.5 ng/mL to about 4 mg/mL, of about 0.5 ng/mL to about 3.5 mg/mL, of about 0.5 ng/mL to about 3 mg/mL, of about 0.5 ng/mL to about 2.5 mg/mL, of about 0.5 ng/mL to about 2 mg/mL, of about 1 ng/mL to about 2 mg/mL, of about 1 ng/mL to about 1.5 mg/mL, of about 100 ng/mL to about 1.1 mg/mL, of about 100 ng/mL to about 1 mg/mL, of about 100 ng/mL to about 500 ng/mL, of about 400 ng/mL to about 1.1 mg/mL, of about 350 ng/mL to about 1.1 mg/mL, of about 300 ng/mL to about 1.1 mg/mL, of about 250 ng/mL to about 1.1 mg/mL, or of about 200 ng/mL to about 1.1 mg/mL, in vitro, optionally as measured by any of the neutralization assays described in the Examples herein. In another embodiment, the isolated antibody or antigen-binding antibody fragment may neutralize SARS-CoV and/or SARS-CoV-2 at an IC90 of about 10 μg/mL or less, an IC90 of about 1 to about 10 μg/mL, an IC90 of about 2 to about 10 μg/mL, an IC90 of about 3 to about 10 μg/mL, an IC90 of about 5 to 10 μg/mL, an IC90 of about 7 to 10 μg/mL, an IC90 of about 8 to 10 μg/mL, an IC90 of about 9 to 10 μg/mL, an IC90 of about 10 μg/mL, 9 μg/mL, 8 μg/mL, 7, μg/mL, 6 μg/mL, or 5 μg/mL.
[0098] In further embodiments, the isolated antibody or antigen-binding antibody fragment may bind to CoV-S (S protein of any CoV, such as but not limited to SARS-CoV-S and/or SARS-CoV -2-S) with a KD value of: (i) 100 nM or lower; (ii) 10 nM or lower; (iii) 1 nM or lower; (iv) 100 pM or lower; (v) 10 pM or lower; (vi) 1 pM or lower; or (vii) 0.1 pM or lower.
[0099] In some embodiments, the antibody, or antigen-binding fragment thereof, is administered intravenously. In other embodiments, the antibody, or antigen-binding fragment thereof, is administered intramuscularly.
[0100] In some embodiments, at least one of the antibody, or antigen-binding fragment thereof, is administered. In some embodiments, at least two of the antibody, or antigen-binding fragment thereof, are administered. In some embodiments, the anti-CoV-S antibody and antigen-binding fragment thereof, e.g., ADI-58125, may be used in combination with a second antibody, or antigen-binding fragment thereof, wherein the second antibody, or antigen-binding fragment thereof, is selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, ADI-58131, or a combination thereof. In some embodiment, the second antibody, or antigen-binding fragment thereof, is ADI-58122. In one embodiment, the second antibody, or antigen-binding fragment thereof, is ADI-58127. In one embodiment, the second antibody, or antigen-binding fragment thereof, is ADI-58129. In one embodiment, the second antibody, or antigen-binding fragment thereof, is ADI-58131.
[0101] In some embodiments, the antibody, or antigen-binding fragment thereof, is administered at a dose of about 100 mg to about 5000 mg, about 100 mg to about 4500 mg, about 100 mg to about 4000 mg, about 100 mg to about 3500 mg, about 100 mg to about 3000 mg, about 100 mg to about 2500 mg, about 300 mg to about 4500 mg, about 500 mg to about 4500 mg, about 600 mg to about 4500 mg, about 1200 mg to about 4500 mg, about 100 mg to about 2000 mg, about 200 mg to about 1500 mg, about 300 mg to about 600 mg, about 500 mg to about 1200 mg, or about 300 mg to about 1200 mg. In some embodiments, the antibody, or antigen-binding fragment thereof, is administered at a dose of about 150 mg, about 200 mg, about 300 mg, about 400 mg, about 450 mg, about 500 mg, about 600 mg, about 700 mg, about 800 mg, about 900 mg, about 1000 mg about 1100 mg, about 1200 mg, about 1300 mg, about 1400 mg, about 1500 mg, about 1600 mg, about 1700 mg, about 1800 mg, about 1900 mg, about 2000 mg, about 2500 mg, about 3000 mg, about 3500 mg, about 4000 mg, about 4500 mg, or about 5000 mg.
[0102] In some embodiments, the antibody, or antigen-binding fragment thereof, is administered at a dose of about 150 mg intramuscularly, about 300 mg intramuscularly, about 450 mg intramuscularly, about 500 mg intravenously, about 600 mg intramuscularly, about 1200 mg intramuscularly, about 1200 mg intravenously, about 4500 mg intramuscularly, or about 4500 mg intravenously.
[0103] In some embodiments, the antibody, or antigen-binding fragment thereof, is administered at a dosage determined based on the current coronavirus variant(s) that is (are) currently circulating in the population (also referred to herein as “variant-based dosing”). For example, in one embodiment, the antibody, or antigen-binding fragment thereof, is administered intramuscularly as a single 1200 mg dose. In another embodiment, the antibody, or antigen-binding fragment thereof, is administered intravenously as a 1200 mg dose. In another embodiment, the antibody, or antigen-binding fragment thereof, is administered intravenously as a single 600 mg dose. In another embodiment, the antibody, or antigen-binding fragment thereof, is administered intravenously as a single 500 mg dose. In another embodiment, the antibody, or antigen-binding fragment thereof, is administered intramuscularly as two 600 mg doses (for example, a first 600 mg dose in the right thigh or right arm, and a second 600 mg dose in the left thigh or left arm; or a first 600 mg dose in the left thigh or left arm, and a second 600 mg dose in the right thigh or right arm) on the same day, for a total dosage of 1200 mg. In another embodiment, the antibody, or antigen-binding fragment thereof, is administered intramuscularly as two 300 mg doses (for example, a first 300 mg dose in the right thigh or right arm, and a second 300 mg dose in the left thigh or left arm; or a first 300 mg dose in the left thigh or left arm, and a second 300 mg dose in the right thigh or right arm) on the same day, for a total dosage of 600 mg.
[0104] In one embodiment, the antibody, or antigen-binding fragment thereof, is administered once. In one embodiment, the antibody, or antigen-binding fragment thereof, is administered weekly. In another embodiment, the antibody, or antigen-binding fragment thereof, is administered daily,
weekly, every two weeks, monthly, or every two months. In one embodiment, the antibody, or antigen-binding fragment thereof, is administered weekly for about four weeks, once weekly for about a month, weekly for about 5 weeks, weekly for about 6 weeks, weekly for about 7 weeks, or weekly for about two months.
[0105] In one embodiment, the methods further comprise obtaining a serum sample from the subject. [0106] In some embodiments, the antibody, or antigen-binding fragment thereof, reaches a maximum concentration (Cmax) of about 10 μg/mL to about 1000 μg/mL, about 20 μg/mL to about 500 μg/mL, about 30 μg/mL to about 400 μg/mL, about 40 μg/mL to about 300 μg/mL, about 50 μg/mL to about 200 μg/mL, about 30 μg/mL to about 200 μg/mL, about 100 μg/mL to about 200 μg/mL, about 50 μg/mL to about 100 μg/mL, or about 30 μg/mL to about 70 μg/mL, about 100 μg/mL to about 400 μg/mL, or about 150 μg/mL to about 350 μg/mL in the serum sample of the subject. In some embodiments, the antibody, or antigen-binding fragment thereof, reaches a maximum concentration (Cmax) of about 30 μg/mL, about 40 μg/mL, about 50 μg/mL, about 60 μg/mL, about 70 μg/mL, about 80 μg/mL, about 90 μg/mL, about 100 μg/mL, about 110 μg/mL, about 120 μg/mL, about 130 μg/mL, about 140 μg/mL, about 150 μg/mL, about 160 μg/mL, about 170 μg/mL, about 180 μg/mL, about 190 μg/mL, or about 200 μg/mL, about 210 μg/mL, about 220 μg/mL, about 230 μg/mL, about 240 μg/mL, about 250 μg/mL, about 260 μg/mL, about 270 μg/mL, about 280 μg/mL, about 290 μg/mL, about 300 μg/mL, about 350 μg/mL, or about 400 μg/mL in the serum sample of the subject. [0107] In some embodiments, the median time for the antibody, or antigen-binding fragment thereof, to reach a maximum concentration in the serum sample of the subject is about 0.01-30 days, about 0.01-0.05 days, about 5-30 days, about 3-20 days, about 6-20 days, about 7-18 days, or about 8-15 days, or about 13-15 days after administration. In some embodiments, the median time for the antibody, or antigen-binding fragment thereof, to reach a maximum concentration in the serum sample of the subject is about 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 days after administration. In some embodiments, the median time for the antibody, or antigen-binding fragment thereof, to reach a maximum concentration in the serum sample of the subject is less than about 1 day, e.g., about 0.01, about 0.02, about 0.03, about 0.04, about 0.05, about 0.06, about 0.07, about 0.08, about 0.09, about 0.1, about 0.2, about 0.3, about 0.4, about 0.5, about 0.6, about 0.7, about 0.8, or about 0.9 days after administration.
[0108] In some embodiments, the antibody, or antigen-binding fragment thereof, reaches a maximum concentration (Cmax) of about 30 μg/mL to about 200 μg/mL, about 100 μg/mL to about 200 μg/mL, about 30 μg/mL to about 100 μg/mL, about 40 μg/mL to about 80 μg/mL, about 50 μg/mL to about 70 μg/mL, or about 30 μg/mL to about 65 μg/mL in the serum sample of the subject in about about 0.01-20 days, about 3-20 days, about 6-20 days, about 7-18 days, about 8-15 or about 13-15 days after administration.
[0109] In some embodiments, the area under the serum concentration-time curve from day 0 to day 21 (AUCo -2i d) is about 100-2000 day* mg/mL, about 1000-2000 day* mg/mL, about 1400-2000 day* mg/mL, about 200-1500 day* mg/mL, about 400-1400 day* mg/mL, about 500-1300 day* mg/mL, about 600-1000 day* mg/mL, or about 800-900 day* mg/mL.
[0110] In some embodiments, the area under the serum concentration-time curve from day 0 to day 90 (AUCo 90d) is about 1000-10000 day* mg/mL, about 2000-8000 day* mg/mL, about 2000-5000 day* mg/mL, about 3000-4000 day* mg/mL, about 5000-10000 day* mg/mL, about 2000-4000 day* mg/mL, about 5000-8000 day* mg/mL, or about 6000-8000 day* mg/mL.
[0111] In some embodiments, the area under the serum concentration-time curve from day 0 to day 180 (AUCo i80d) is about 1000-10000 day* mg/mL, about 2000-5000 day* mg/mL, about 3000-6000 day* mg/mL, about 5000-10000 day* mg/mL, about 5000-8000 day* mg/mL, or about 4000-6000 day* mg/mL.
[0112] In some embodiments, the area under the serum concentration-time curve from day 0 to day 365 (AUCo 365d) is about 1000-30000 day* mg/mL, about 5000-30000 day* mg/mL, about 10000- 30000 day* mg/mL, about 10000-15000 day* mg/mL, about 20000-30000 day* mg/mL, or about 25000-30000 day* mg/mL.
[0113] In some embodiments, the antibody, or antigen-binding fragment thereof, has a virus neutralizing titer of about 100-2000, about 200-1500, about 300-1500, or about 500-1500 in the serum sample of the subject about 6 months after administration. In some embodiments, the antibody, or antigen-binding fragment thereof, has a virus neutralizing titer of about 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, 1100, 1200, 1300, 1400, 1500, 1600, 1700, 1800, 1900 or 2000 in the serum sample of the subject about 6 months after administration.
[0114] In some embodiments, the antibody, or antigen-binding fragment thereof, has a serum half- life of about 50-200 days, about 50-160 days, about 50-140 days, about 40-130 days, about 50-100 days, about 60-100 days, about 60-90 days, about 70-90 days, about 90-100 days, about 70-80 days, about 80-130 days, about 100-150 days, about 110-140 days, or about 120-140 days.
[0115] In some embodiments, the method further comprises obtaining an epithelial lining fluid (ELF) sample from the subject. In some embodiments, the ELF sample comprises an ELF sample from an upper airway, an lower airway, and/or an alveolar tissue.
[0116] In some embodiments, the antibody, or antigen-binding fragment thereof, reaches a concentrationof about 1 μg/mL to about 100 μg/mL, about 1 μg/mL to about 80 μg/mL, about 80 μg/mL to about 100 μg/mL, about 50 μg/mL to about 100 μg/mL, about 1 μg/mL to about 50 μg/mL, about 2 μg/mL to about 25 μg/mL, or about 2 μg/mL to about 10 μg/mL in the ELF sample of the subject.
[0117] In some embodiments, the antibody, or antigen-binding fragment thereof, has a receptor occupancy of at least 50%, 60%, 70%, 80%, or 90% in the ELF sample. In some embodiments, the
antibody, or antigen-binding ragment thereof, has a receptor occupancy of at least 50%, 60%, 70%, 80%, or 90% in the ELF sample for about 28 days after administration.
[0118] In some embodiments, the 80% virus neutralization titer (MN80) of the antibody, or antigen binding fragment thereof, at about days 0-14, about days 7-21, about 3 months, or about 6 months after administration, is about 10-6000, about 50-600, about 500-1500, about 1000-2500, about 100- 2500, about 500-2000, about 500-1500, about 400-1200, about 200-1500, about 300-1000, about 400- 800, about 400-1000, or about 500-600 in the subject.
[0119] In some embodiments, the 80% virus neutralization titer (MN80) of the antibody, or antigen binding fragment thereof, at about day 7, day 14, or day 21 after administration, is about 300-2000, about 400-600, about 600-1500, about 1100-1700, about 500-1700, about 500-1500, about 400-1200, about 400-800, about 400-1000, or about 500-600 in the serum sample of the subject.
[0120] In some embodiments, the 80% virus neutralization titer (MN80) of the antibody, or antigen binding fragment thereof, at about 3 months after administration, is about 200-1000, about 200-800, about 200-500, about 400-900, or about 400-600 in the serum sample of the subject.
[0121] In some embodiments, the 80% virus neutralization titer (MN80) of the antibody, or antigen binding fragment thereof, at about 6 months after administration, is about 10-500, about 300-500, or about 50-200 in the serum sample of the subject.
[0122] In some embodiments, the 50% virus neutralization titer (MN50) of the antibody, or antigen binding fragment thereof, at about days 0-14, about days 7-21, about 3 months, about 6 months, or about 12 months after administration, is about 100-6000, about 300-1500, about 1700-3800, about 3800-5200, about 300-5500, about 1200-4500, about 1300-4300, about 1200-4000, about 100-2500, about 500-2500, about 800-2000, about 1000-1800, about 800-1300, about 900-1100, or about 1300- 1500 in the serum sample of the subject.
[0123] In some embodiments, the 50% virus neutralization titer (MN50) of the antibody, or antigen binding fragment thereof, at about day 7, day 14 or day 21 after administration, is about 1000-4500, about 1200-4500, about 1300-4300, about 1200-3900, about 1500-4000, about 1800-3800, about 3800-4500, about 1000-1800 or about 1200-1500 in the serum sample of the subject.
[0124] In some embodiments, the 50% virus neutralization titer (MN50) of the antibody, or antigen binding fragment thereof, at about 3 months after administration, is about 800-1300 or about 900- 1100 in the serum sample of the subject.
[0125] In some embodiments, the 50% virus neutralization titer (MN50) of the antibody, or antigen binding fragment thereof, at about 6 months after administration, is about 200-500 or about 300-600 in the serum sample of the subject.
[0126] In some embodiments, the 50% virus neutralization titer (MN50) of the antibody, or antigen binding fragment thereof, at about 12 months after administration, is about 150-500 or about 200-400 in the serum sample of the subject.
[0127] In some embodiments, the neutralization titer is determined using a plaque reduction neutralization test (PRNT).
[0128] In some embodiments, the serum concentration of the antibody, or antigen-binding fragment thereof, is about 1-300 mg/L, about 1-250 mg/L, about 1-200 mg/L, about 1-100 mg/L, about 100-250 mg/L, about 150-200 mg/L, about 120-170 mg/L, about 1-90 mg/L, about 1-80 mg/L, about 1-70 mg/L, about 1-60 mg/L, about 1-50 mg/L, about 1-40 mg/L, about 1-30 mg/L, about 1-20 mg/L, about 1-10 mg/L, about 2-9 mg/L, about 3-8 mg/L, about 4-6 mg/L, about 5-80 mg/L, about 10-70 mg/L, or about 10-60 mg/L in the subject about 3 months after administration.
[0129] In some embodiments, the serum concentration of the antibody, or antigen-binding fragment thereof, is about 1 mg/L, about 2 mg/L, about 3 mg/L, about 4 mg/L, about 5 mg/L, about 6 mg/L, about 7 mg/L, about 8 mg/L, about 9 mg/L, about 10 mg/L, about 11 mg/L, about 12 mg/L, about 13 mg/L, about 14 mg/L, about 15 mg/L, about 16 mg/L, about 17 mg/L, about 18 mg/L, about 19 mg/L, about 20 mg/L, about 25 mg/L, about 30 mg/L, about 35 mg/L, about 40 mg/L, about 45 mg/L, about 50 mg/L, about 55 mg/L, about 60 mg/L, about 65 mg/L, about 70 mg/L, about 75 mg/L, about 80 mg/L, about 85 mg/L, about 90 mg/L, about 95 mg/L, about 100 mg/L, about 110 mg/L, about 120 mg/L, about 130 mg/L, about 140 mg/L, about 150 mg/L, about 160 mg/L, about 170 mg/L, about 180 mg/L, about 190 mg/L, about 200 mg/L, about 210 mg/L, about 220 mg/L, about 230 mg/L, about 240 mg/L, about 250 mg/L, about 300 mg/L in the subject about 3 months after administration. [0130] In some embodiments, the serum concentration of the antibody, or antigen-binding fragment thereof, is about 1-100 mg/L, about 1-90 mg/L, about 1-80 mg/L, about 1-70 mg/L, about 1-60 mg/L, about 1-50 mg/L, about 1-40 mg/L, about 1-30 mg/L, about 1-20 mg/L, about 1-10 mg/L, about 2-9 mg/L, about 3-8 mg/L, about 4-6 mg/L, about 5-80 mg/L, about 10-70 mg/L, about 10-60 mg/L, about 5-25 mg/L, about 10-30 mg/L, about 15-25 mg/L, about 20-100 mg/L, about 30-100 mg/L, about 50- 80 mg/L, or about 60-70 mg/L in the subject about 6 months after administration.
[0131] In some embodiments, the serum concentration of the antibody, or antigen-binding fragment thereof, is about 1 mg/L, about 2 mg/L, about 3 mg/L, about 4 mg/L, about 5 mg/L, about 6 mg/L, about 7 mg/L, about 8 mg/L, about 9 mg/L, about 10 mg/L, about 11 mg/L, about 12 mg/L, about 13 mg/L, about 14 mg/L, about 15 mg/L, about 16 mg/L, about 17 mg/L, about 18 mg/L, about 19 mg/L, about 20 mg/L, about 25 mg/L, about 30 mg/L, about 35 mg/L, about 40 mg/L, about 45 mg/L, about 50 mg/L, about 55 mg/L, about 60 mg/L, about 65 mg/L, about 70 mg/L, about 75 mg/L, about 80 mg/L, about 85 mg/L, about 90 mg/L, about 95 mg/L, or about 100 mg/L in the subject about 6 months after administration.
[0132] In some embodiments, the serum concentration of the antibody, or antigen-binding fragment thereof, is about 0.1-30 mg/L, about 1-30 mg/L, about 1-20 mg/L, about 1-10 mg/L, about 10-20 mg/L, about 0.1-3 mg/L, about 0.5-8 mg/L, about 0.5-10 mg/L, about 2-9 mg/L, about 5-15 mg/L, about 3-8 mg/L, or about 4-6 mg/L in the subject about 12 months after administration.
[0133] In some embodiments, the serum concentration of the antibody, or antigen-binding fragment thereof, is about 0.1 mg/L, about 0.2 mg/L, about 0.3 mg/L, about 0.4 mg/L, about 0.5 mg/L, about 0.6 mg/L, about 0.7 mg/L, about 0.8 mg/L, about 0.9 mg/L, about 1 mg/L, about 2 mg/L, about 3 mg/L, about 4 mg/L, about 5 mg/L, about 6 mg/L, about 7 mg/L, about 8 mg/L, about 9 mg/L, about 10 mg/L, about 15 mg/L, about 20 mg/L, about 25 mg/L, or about 30 mg/L in the subject about 12 months after administration.
[0134] In some embodiments, the clearance rate for the antibody, or antigen-binding fragment thereof, is about 0.1-10 mL/d, about 0.1-5.0 mL/d, about 0.1-3.0 mL/d, about 0.5-2.5 mL/d, or about 1.0-2.0 mL/d.
[0135] In some embodiments, the steady state volume of distribution for the antibody, or antigen binding fragment thereof, is about 1-10 L, about 2-8 L, about 4-9 L, about 4-8 L, or about 5-7L.
[0136] In some embodiments, administration of the antibody, or antigen-binding fragment thereof, reduces pulmonary inflammation in the subject.
[0137] In some embodiments, administration of the antibody, or antigen-binding fragment thereof, reduces the risk of COVID-19 hospitalization or death of the subject. In some embodiments, the risk of COVID-19 hospitalization or death is reduced by at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90% or 95%.
[0138] In some embodiments, administration of the antibody, or antigen-binding fragment thereof, reduces viral load in the subject. In some embodiments, the viral load is reduced by at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90% or 95%. In some embodiments, the viral load is reduced by at least about 0.5 loglO copies/mL, at least about 0.6 loglO copies/mL, at least about 0.7 loglO copies/mL, at least about 0.8 loglO copies/mL, at least about 0.9 loglO copies/mL, at least about 1.0 loglO copies/mL, at least about 1.1 loglO copies/mL, at least about 1.2 loglO copies/mL, at least about 1.3 loglO copies/mL, at least about 1.4 loglO copies/mL, at least about 1.5 loglO copies/mL, at least about 1.6 loglO copies/mL, at least about 1.7 loglO copies/mL, at least about 1.8 loglO copies/mL, at least about 1.9 loglO copies/mL, at least about 2.0 loglO copies/mL, at least about 2.1 loglO copies/mL, at least about 2.2 loglO copies/mL, at least about 2.3 loglO copies/mL, at least about 2.4 loglO copies/mL, at least about 2.5 loglO copies/mL, at least about 2.6 loglO copies/mL, at least about 2.7 loglO copies/mL, at least about 2.8 loglO copies/mL, at least about 2.9 loglO copies/mL, or at least about 3.0 loglO copies/mL.
[0139] In one aspect, the present disclosure also relates to kits comprising: (a) at least one isolated antibody or antigen-binding antibody fragment disclosed herein; and (b) an instruction for using the antibody or antigen-binding antibody fragment.
[0140] In some embodiments, the kit may be for use in: (i) determining whether a CoV is present in a subject; (ii) diagnosing whether a subject has CoV infection; (iii) predicting whether a CoV vaccine
will elicit a protective immune response; or (iv) predicting whether a CoV vaccine will elicit a neutralizing antibody response.
[0141] In one aspect, the present disclosure provides a method of inducing an immune response against a coronavirus (CoV) in a subject in need thereof, the method comprising administering to the subject an isolated antibody, or antigen-binding fragment thereof, which binds to the spike protein of a coronavirus (“CoV-S”), wherein said antibody, or antigen-binding fragment thereof, comprises a heavy chain variable region (VH) comprising a VH CDR1 comprising SEQ ID NO:52, a VH CDR2 comprising SEQ ID NO:54, and a VH CDR3 comprising SEQ ID NO:56, and a light chain variable region (VL) comprising a VL CDR1 comprising SEQ ID NO:252, a VL CDR2 comprising SEQ ID NO:254, and a VL CDR3 comprising SEQ ID NO:256; (a) wherein the antibody, or antigen-binding fragment thereof, reaches a maximum concentration (Cmax) of about 10 μg/mL to about 1000 μg/mL, about 20 μg/mL to about 500 μg/mL, about 30 μg/mL to about 400 μg/mL, about 40 μg/mL to about 300 μg/mL, about 50 μg/mL to about 200 μg/mL, about 50 μg/mL to about 100 μg/mL, or about 30 μg/mL to about 70 μg/mL, about 100 μg/mL to about 400 μg/mL, about 150 μg/mL to about 350 μg/mL in serum of the subject; (b) wherein the median time (Tmax) for the antibody, or antigen binding fragment thereof, to reach a maximum concentration in serum of the subject is about 0.01-30 days, about 3-20 days, about 6-20 days, about 7-18 days, about 8-15 or about 13-15 days; (c) wherein the antibody, or antigen-binding fragment thereof, reaches a maximum concentration of about 30 μg/mL to about 200 μg/mL, about 100 μg/mL to about 200 μg/mL, about 30 μg/mL to about 100 μg/mL, about 40 μg/mL to about 80 μg/mL, about 50 μg/mL to about 70 μg/mL, or about 30 μg/mL to about 65 μg/mL in serum of the subject in about 0.01-20 days, about 3-20 days, about 6-20 days, about 7-18 days, about 8-15 or about 13-15 days after administration; (d) wherein the area under the serum concentration-time curve from day 0 to day 21 (AUC0-21d) is about 100-2000 day* μg/mL, about 1000-2000 day* μg/mL, about 1400-2000 day* μg/mL, about 200-1500 day* μg/mL, about 400-1400 day* μg/mL, about 500-1300 day* μg/mL, about 600-1000 day* μg/mL, or about 800-900 day* μg/mL; (e) wherein the area under the serum concentration-time curve from day 0 to day 90 (AUC0-90d) is about 1000-10000 day* μg/mL, about 2000-8000 day* μg/mL, about 2000-5000 day* μg/mL, about 3000-4000 day* μg/mL, about 5000-10000 day* μg/mL, about 2000-4000 day* μg/mL, about 5000-8000 day* μg/mL, or about 6000-8000 day* μg/mL; (f) wherein the area under the serum concentration-time curve from day 0 to day 180 (AUC0-180d) is about 1000-10000 day* μg/mL, about 2000-5000 day* μg/mL, about 3000-6000 day* μg/mL, about 5000-10000 day* μg/mL, about 5000-8000 day* μg/mL, or about 4000-6000 day* μg/mL; (g) wherein the area under the serum concentration-time curve from day 0 to day 365 (AUC0-365d) is about 1000-30000 day* μg/mL, about 5000-30000 day* μg/mL, about 10000-30000 day* μg/mL, about 10000-15000 day* μg/mL, about 20000-30000 day* μg/mL, or about 25000-30000 day* μg/mL; and/or (h) wherein the antibody, or antigen-binding fragment thereof, has a serum half-life of about 50-200 days, about 50-
160 days, about 50-140 days, about 40-130 days, about 50-100 days, about 60-100 days, about 60-90 days, about 70-90 days, about 90-100 days, about 70-80 days, about 80-130 days, about 100-150 days, about 110-140 days, or about 120-140 days.
[0142] In one aspect, the present disclosure provides a method of preventing a coronavirus infection in a subject in need thereof, the method comprising administering to the subject an isolated antibody, or antigen-binding fragment thereof, which binds to the spike protein of a coronavirus (“CoV-S”), wherein said antibody, or antigen-binding fragment thereof, comprises a heavy chain variable region (VH) comprising a VH CDR1 comprising SEQ ID NO:52, a VH CDR2 comprising SEQ ID NO:54, and a VH CDR3 comprising SEQ ID NO: 56, and a light chain variable region (VL) comprising a VL CDR1 comprising SEQ ID NO:252, a VL CDR2 comprising SEQ ID NO:254, and a VL CDR3 comprising SEQ ID NO:256; (a) wherein the antibody, or antigen-binding fragment thereof, reaches a maximum concentration (Cmax) of about 10 μg/mL to about 1000 μg/mL, about 20 μg/mL to about 500 μg/mL, about 30 μg/mL to about 400 μg/mL, about 40 μg/mL to about 300 μg/mL, about 50 μg/mL to about 200 μg/mL, about 50 μg/mL to about 100 μg/mL, or about 30 μg/mL to about 70 μg/mL, about 100 μg/mL to about 400 μg/mL, about 150 μg/mL to about 350 μg/mL in serum of the subject; (b) wherein the median time (Tmax) for the antibody, or antigen-binding fragment thereof, to reach a maximum concentration in serum of the subject is about 0.01-30 days, about 3-20 days, about 6-20 days, about 7-18 days, about 8-15 or about 13-15 days; (c) wherein the antibody, or antigen binding fragment thereof, reaches a maximum concentration of about 30 μg/mL to about 200 μg/mL, about 100 μg/mL to about 200 μg/mL, about 30 μg/mL to about 100 μg/mL, about 40 μg/mL to about 80 μg/mL, about 50 μg/mL to about 70 μg/mL, or about 30 μg/mL to about 65 μg/mL in serum of the subject in about 0.01-20 days, about 3-20 days, about 6-20 days, about 7-18 days, about 8-15 or about 13-15 days after administration; (d) wherein the area under the serum concentration-time curve from day 0 to day 21 (AUC0-21d) is about 100-2000 day* μg/mL, about 1000-2000 day* μg/mL, about 1400-2000 day* μg/mL, about 200-1500 day* μg/mL, about 400-1400 day* μg/mL, about 500-1300 day* μg/mL, about 600-1000 day* μg/mL, or about 800-900 day* μg/mL; (e) wherein the area under the serum concentration-time curve from day 0 to day 90 (AUC0-90d) is about 1000-10000 day* μg/mL, about 2000-8000 day* μg/mL, about 2000-5000 day* μg/mL, about 3000-4000 day* μg/mL, about 5000-10000 day* μg/mL, about 2000-4000 day* μg/mL, about 5000-8000 day* μg/mL, or about 6000-8000 day* μg/mL; (f) wherein the area under the serum concentration-time curve from day 0 to day 180 (AUC0-180d) is about 1000-10000 day* μg/mL, about 2000-5000 day* μg/mL, about 3000-6000 day* μg/mL, about 5000-10000 day* μg/mL, about 5000-8000 day* μg/mL, or about 4000-6000 day* μg/mL; (g) wherein the area under the serum concentration-time curve from day 0 to day 365 (AUC0-365d) is about 1000-30000 day* μg/mL, about 5000-30000 day* μg/mL, about 10000-30000 day* μg/mL, about 10000-15000 day* μg/mL, about 20000-30000 day* μg/mL, or about 25000-30000 day* μg/mL; and/or (h) wherein the antibody, or antigen-binding fragment
thereof, has a serum half-life of about 50-200 days, about 50-160 days, about 50-140 days, about 40- 130 days, about 50-100 days, about 60-100 days, about 60-90 days, about 70-90 days, about 90-100 days, about 70-80 days, about 80-130 days, about 100-150 days, about 110-140 days, or about 120- 140 days.
[0143] In one aspect, the present disclosure provides a method of beating a coronavirus infection in a subject in need thereof, the method comprising administering to the subject an isolated antibody, or antigen-binding fragment thereof, which binds to the spike protein of a coronavirus (“CoV-S”), wherein said antibody, or antigen-binding fragment thereof, comprises a heavy chain variable region (VH) comprising a VH CDR1 comprising SEQ ID NO:52, a VH CDR2 comprising SEQ ID NO:54, and a VH CDR3 comprising SEQ ID NO: 56, and a light chain variable region (VL) comprising a VL CDR1 comprising SEQ ID NO:252, a VL CDR2 comprising SEQ ID NO:254, and a VL CDR3 comprising SEQ ID NO:256; (a) wherein the antibody, or antigen-binding fragment thereof, reaches a maximum concentration (Cmax) of about 10 μg/mL to about 1000 μg/mL, about 20 μg/mL to about 500 μg/mL, about 30 μg/mL to about 400 μg/mL, about 40 μg/mL to about 300 μg/mL, about 50 μg/mL to about 200 μg/mL, about 50 μg/mL to about 100 μg/mL, or about 30 μg/mL to about 70 μg/mL, about 100 μg/mL to about 400 μg/mL, about 150 μg/mL to about 350 μg/mL in serum of the subject; (b) wherein the median time (Tmax) for the antibody, or antigen-binding fragment thereof, to reach a maximum concentration in serum of the subject is about 0.01-30 days, about 3-20 days, about 6-20 days, about 7-18 days, about 8-15 or about 13-15 days; (c) wherein the antibody, or antigen binding fragment thereof, reaches a maximum concentration of about 30 μg/mL to about 200 μg/mL, about 100 μg/mL to about 200 μg/mL, about 30 μg/mL to about 100 μg/mL, about 40 μg/mL to about 80 μg/mL, about 50 μg/mL to about 70 μg/mL, or about 30 μg/mL to about 65 μg/mL in serum of the subject in about 0.01-20 days, about 3-20 days, about 6-20 days, about 7-18 days, about 8-15 or about 13-15 days after administration; (d) wherein the area under the serum concentration-time curve from day 0 to day 21 (AUC0-21d) is about 100-2000 day* μg/mL, about 1000-2000 day* μg/mL, about 1400-2000 day* μg/mL, about 200-1500 day* μg/mL, about 400-1400 day* μg/mL, about 500-1300 day* μg/mL, about 600-1000 day* μg/mL, or about 800-900 day* μg/mL; (e) wherein the area under the serum concentration-time curve from day 0 to day 90 (AUC0-90d) is about 1000-10000 day* μg/mL, about 2000-8000 day* μg/mL, about 2000-5000 day* μg/mL, about 3000-4000 day* μg/mL, about 5000-10000 day* μg/mL, about 2000-4000 day* μg/mL, about 5000-8000 day* μg/mL, or about 6000-8000 day* μg/mL; (f) wherein the area under the serum concentration-time curve from day 0 to day 180 (AUC0-180d) is about 1000-10000 day* μg/mL, about 2000-5000 day* μg/mL, about 3000-6000 day* μg/mL, about 5000-10000 day* μg/mL, about 5000-8000 day* μg/mL, or about 4000-6000 day* μg/mL; (g) wherein the area under the serum concentration-time curve from day 0 to day 365 (AUC0-365d) is about 1000-30000 day* μg/mL, about 5000-30000 day* μg/mL, about 10000-30000 day* μg/mL, about 10000-15000 day* μg/mL, about 20000-30000 day* μg/mL,
or about 25000-30000 day* mg/mL; and/or (h) wherein the antibody, or antigen-binding fragment thereof, has a serum half-life of about 50-200 days, about 50-160 days, about 50-140 days, about 40- 130 days, about 50-100 days, about 60-100 days, about 60-90 days, about 70-90 days, about 90-100 days, about 70-80 days, about 80-130 days, about 100-150 days, about 110-140 days, or about 120- 140 days.
[0144] In one aspect, the present disclosure provides a method of treating a symptom of a coronavirus infection in a subject in need thereof, the method comprising administering to the subject an isolated antibody, or antigen-binding fragment thereof, which binds to the spike protein of a coronavirus (“CoV-S”), wherein said antibody, or antigen-binding fragment thereof, comprises a heavy chain variable region (VH) comprising a VH CDR1 comprising SEQ ID NO:52, a VH CDR2 comprising SEQ ID NO:54, and a VH CDR3 comprising SEQ ID NO:56, and a light chain variable region (VL) comprising a VL CDR1 comprising SEQ ID NO:252, a VL CDR2 comprising SEQ ID NO:254, and a VL CDR3 comprising SEQ ID NO:256; (a) wherein the antibody, or antigen-binding fragment thereof, reaches a maximum concentration (Cmax) of about 10 μg/mL to about 1000 μg/mL, about 20 μg/mL to about 500 μg/mL, about 30 μg/mL to about 400 μg/mL, about 40 μg/mL to about 300 μg/mL, about 50 μg/mL to about 200 μg/mL, about 50 μg/mL to about 100 μg/mL, or about 30 μg/mL to about 70 μg/mL, about 100 μg/mL to about 400 μg/mL, about 150 μg/mL to about 350 μg/mL in serum of the subject; (b) wherein the median time (Tmax) for the antibody, or antigen binding fragment thereof, to reach a maximum concentration in serum of the subject is about 0.01-30 days, about 3-20 days, about 6-20 days, about 7-18 days, about 8-15 or about 13-15 days; (c) wherein the antibody, or antigen-binding fragment thereof, reaches a maximum concentration of about 30 μg/mL to about 200 μg/mL, about 100 μg/mL to about 200 μg/mL, about 30 μg/mL to about 100 μg/mL, about 40 μg/mL to about 80 μg/mL, about 50 μg/mL to about 70 μg/mL, or about 30 μg/mL to about 65 μg/mL in serum of the subject in about 0.01-20 days, about 3-20 days, about 6-20 days, about 7-18 days, about 8-15 or about 13-15 days after administration; (d) wherein the area under the serum concentration-time curve from day 0 to day 21 (AUC0-21d) is about 100-2000 day* μg/mL, about 1000-2000 day* μg/mL, about 1400-2000 day* μg/mL, about 200-1500 day* μg/mL, about 400-1400 day* μg/mL, about 500-1300 day* μg/mL, about 600-1000 day* μg/mL, or about 800-900 day* μg/mL; (e) wherein the area under the serum concentration-time curve from day 0 to day 90 (AUC0-90d) is about 1000-10000 day* μg/mL, about 2000-8000 day* μg/mL, about 2000-5000 day* μg/mL, about 3000-4000 day* μg/mL, about 5000-10000 day* μg/mL, about 2000-4000 day* μg/mL, about 5000-8000 day* μg/mL, or about 6000-8000 day* μg/mL; (f) wherein the area under the serum concentration-time curve from day 0 to day 180 (AUC0-180d) is about 1000-10000 day* μg/mL, about 2000-5000 day* μg/mL, about 3000-6000 day* μg/mL, about 5000-10000 day* μg/mL, about 5000-8000 day* μg/mL, or about 4000-6000 day* μg/mL; (g) wherein the area under the serum concentration-time curve from day 0 to day 365 (AUC0-365d) is about 1000-30000 day* μg/mL,
about 5000-30000 day* mg/mL, about 10000-30000 day* mg/mL, about 10000-15000 day* mg/mL, about 20000-30000 day* mg/mL, or about 25000-30000 day* mg/mL; and/or (h) wherein the antibody, or antigen-binding fragment thereof, has a serum half-life of about 50-200 days, about 50-160 days, about 50-140 days, about 40-130 days, about 50-100 days, about 60-100 days, about 60-90 days, about 70-90 days, about 90-100 days, about 70-80 days, about 80-130 days, about 100-150 days, about 110-140 days, or about 120-140 days.
[0145] In some embodiments, the symptom comprises at least one of bronchitis, pneumonia, respiratory failure, acute respiratory failure, organ failure, multi-organ system failure, pediatric inflammatory multisystem syndrome, acute respiratory distress syndrome, blood clot, a cardiac condition, myocardial injury, myocarditis, heart failure, cardiac arrest, acute myocardial infarction, dysrhythmia, venous thromboembolism, post-intensive care syndrome, shock, anaphylactic shock, cytokine release syndrome, septic shock, disseminated intravascular coagulation, ischemic stroke, intracerebral hemorrhage, microangiopathic thrombosis, psychosis, seizure, nonconvulsive status epilepticus, traumatic brain injury, stroke, anoxic brain injury, encephalitis, posterior reversible leukoencephalopathy, necrotizing encephalopathy, post-infectious encephalitis, autoimmune mediated encephalitis, acute disseminated encephalomyelitis, acute kidney injury, acute liver injury, pancreatic injury, immune thrombocytopenia, subacute thyroiditis, a gastrointestinal complication, aspergillosis, increased susceptibility to infection with another virus or bacteria, and/or a pregnancy-related complication.
[0146] In one aspect, the present disclosure provides a method of decreasing the risk of mortality, hospitalization, mechanical ventilation, or a combination thereof, in a subject infected by a coronavirus, the method comprising administering to the subject an isolated antibody, or antigen binding fragment thereof, which binds to the spike protein of a coronavirus (“CoV-S”), wherein said antibody, or antigen-binding fragment thereof, comprises a heavy chain variable region (VH) comprising a VH CDR1 comprising SEQ ID NO:52, a VH CDR2 comprising SEQ ID NO:54, and a VH CDR3 comprising SEQ ID NO: 56, and a light chain variable region (VL) comprising a VL CDR1 comprising SEQ ID NO:252, a VL CDR2 comprising SEQ ID NO:254, and a VL CDR3 comprising SEQ ID NO:256; (a) wherein the antibody, or antigen-binding fragment thereof, reaches a maximum concentration (Cmax) of about 10 μg/mL to about 1000 μg/mL, about 20 μg/mL to about 500 μg/mL, about 30 μg/mL to about 400 μg/mL, about 40 μg/mL to about 300 μg/mL, about 50 μg/mL to about 200 μg/mL, about 50 μg/mL to about 100 μg/mL, or about 30 μg/mL to about 70 μg/mL, about 100 μg/mL to about 400 μg/mL, about 150 μg/mL to about 350 μg/mL in serum of the subject; (b) wherein the median time (Tmax) for the antibody, or antigen-binding fragment thereof, to reach a maximum concentration in serum of the subject is about 0.01-30 days, about 3-20 days, about 6-20 days, about 7-18 days, about 8-15 or about 13-15 days; (c) wherein the antibody, or antigen binding fragment thereof, reaches a maximum concentration of about 30 μg/mL to about 200 μg/mL,
about 100 mg/mL to about 200 mg/mL, about 30 mg/mL to about 100 mg/mL, about 40 mg/mL to about 80 mg/mL, about 50 mg/mL to about 70 mg/mL, or about 30 mg/mL to about 65 mg/mL in serum of the subject in about 0.01-20 days, about 3-20 days, about 6-20 days, about 7-18 days, about 8-15 or about 13-15 days after administration; (d) wherein the area under the serum concentration-time curve from day 0 to day 21 (AUC0-21d) is about 100-2000 day* mg/mL, about 1000-2000 day* mg/mL, about 1400-2000 day* mg/mL, about 200-1500 day* mg/mL, about 400-1400 day* mg/mL, about 500-1300 day* mg/mL, about 600-1000 day* mg/mL, or about 800-900 day* mg/mL; (e) wherein the area under the serum concentration-time curve from day 0 to day 90 (AUC0-90d) is about 1000-10000 day* mg/mL, about 2000-8000 day* mg/mL, about 2000-5000 day* mg/mL, about 3000-4000 day* mg/mL, about 5000-10000 day* mg/mL, about 2000-4000 day* mg/mL, about 5000-8000 day* mg/mL, or about 6000-8000 day* mg/mL; (f) wherein the area under the serum concentration-time curve from day 0 to day 180 (AUC0-180d) is about 1000-10000 day* mg/mL, about 2000-5000 day* mg/mL, about 3000-6000 day* mg/mL, about 5000-10000 day* mg/mL, about 5000-8000 day* mg/mL, or about 4000-6000 day* mg/mL; (g) wherein the area under the serum concentration-time curve from day 0 to day 365 (AUC0-365d) is about 1000-30000 day* mg/mL, about 5000-30000 day* mg/mL, about 10000-30000 day* mg/mL, about 10000-15000 day* mg/mL, about 20000-30000 day* mg/mL, or about 25000-30000 day* mg/mL; and/or (h) wherein the antibody, or antigen-binding fragment thereof, has a serum half-life of about 50-200 days, about 50-160 days, about 50-140 days, about 40- 130 days, about 50-100 days, about 60-100 days, about 60-90 days, about 70-90 days, about 90-100 days, about 70-80 days, about 80-130 days, about 100-150 days, about 110-140 days, or about 120- 140 days.
[0147] In some embodiments, the CoV-S is the spike protein of SARS-CoV (“SARS-CoV-S”) or the spike protein of SARS-CoV-2 (“SARS-CoV-2-S”).
[0148] In some embodiments, the antibody, or antigen-binding fragment thereof, cross-reacts with SARS-CoV-S and SARS-CoV-2-S.
[0149] In some embodiments, SARS-CoV-S comprises an amino acid sequence of SEQ ID NO: 401, and wherein SARS-CoV-2-S comprises an amino acid sequence of SEQ ID NO: 403.
[0150] In some embodiments, the SARS-CoV -2-S is a B.1.1.7 variant, a B. 1.351 variant, a B.1.1.28 variant, a B. 1.429 variant, a P.l variant, a B.1.617 variant, a B.1.617.2 variant, a C.37 variant, a 1.621 variant, a AY.l variant, a 1.623 variant, a C.36 variant, a A.27 variant, a AV.l variant, a B.1.1.482 variant, a B.1.1.523 variant, a B.1.427 variant, a AY.4 variant, a AY.ll variant, a D614G variant of SEQ ID NO: 403, a B.1.1.529/BA.1 variant, a BA.1.1 variant, or a BA.2 variant.
[0151] In one aspect, the present disclosure provides a method for inducing an immune response against a coronavirus Omicron variant in a subject in need thereof, for treating a coronavirus infection caused by an Omicron variant in a subject in need thereof, for preventing a coronavirus infection caused by an Omicron variant in a subject in need thereof, for treating a symptom of an infection of a
subject by an Omicron variant in a subject in need thereof, and/or for decreasing the risk of mortality, hospitalization, mechanical ventilation, or a combination thereof, in a subject infected by an Omicron variant, the method comprising administering to the subject an isolated antibody, or antigen-binding fragment thereof, wherein said antibody, or antigen-binding fragment thereof, comprises a heavy chain variable region (VH) comprising a VH CDR1 comprising SEQ ID NO:52, a VH CDR2 comprising SEQ ID NO:54, and a VH CDR3 comprising SEQ ID NO:56, and a light chain variable region (VL) comprising a VL CDR1 comprising SEQ ID NO:252, a VL CDR2 comprising SEQ ID NO:254, and a VL CDR3 comprising SEQ ID NO:256.
[0152] In some embodiments, the Omicron variant comprises a B.1.1.529/BA.1 variant, a BA.1.1 variant, or a BA.2 variant.
[0153] In some embodiments, the VH comprises a sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to SEQ ID NO:58 and the VL comprises a sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to SEQ ID NO:258. In some embodiments, the VH comprises or consists of SEQ ID NO:58 and the VL comprises or consists of SEQ ID NO:258.
[0154] In any method described herein, the method may further comprise selecting a subject. In one embodiment, the method may further comprise selecting a subject that would benefit from administration of the antibody, or antigen-binding fragment thereof.
[0155] In some embodiments, the subject is a human subject. In some embodiments, the subject is an adult, an adolescent, or a child.
[0156] In some embodiments, the subject has at least one risk factor which renders them more prone to a poor clinical outcome. In some embodiments, the at least one risk factor is selected from the group consisting of: an old age selected from the group consisting of over 55, over 60 or over 65 years old; diabetes, a chronic respiratory condition, obesity, hypertension, a cardiac or cardiovascular condition, a chronic inflammatory or autoimmune condition, and an immune compromised status. [0157] In some embodiments, the subject is immunocompromised. In some embodiments, the subject is at a high risk of disease progression. In some embodiments, the subject is (a) age 50 years or above with no comorbid conditions or additional risk factors for progression of COVID-19; (b) between about 18 to about 50 years old and with one or more preexisting medical conditions selected from the group consisting of obesity, diabetes, chronic kidney disease, chronic lung disease, cardiac disease, sickle cell disease or thalassemia, solid organ or blood stem cell transplant recipient, other immunodeficiency due to underlying illness or immunosuppressant medication, Down Syndrome, stroke or cerebrovascular disease, substance use disorder, and pregnancy; or (c) between about 12 to about 17 years old and with one or more preexisting medical conditions selected from the group consisting of obesity, diabetes, chronic kidney disease, sickle cell disease or thalassemia, congenital or acquired heart disease, neurodevelopmental disorder, a medically-related technological
dependence, asthma or chornic respiratory disease, solid organ or blood stem cell transplant recipient, other immunodeficiency due to underlying illness or immunosuppressant medication, Down Syndrome, stroke or cerebrovascular disease, substance use disorder, and pregnancy.
[0158] In some embodiments, the subject is age 50 or above with no comorbid conditions or additional risk factors for progression of COVID-19.
[0159] In some embodiments, the subject has hypertension with at least one medication prescribed or recommended.
[0160] In some embodiments, the subject has moderate to severe asthma requiring daily therapy. [0161] In some embodiments, the antibody, or the antigen-binding fragment thereof, is administered intramuscularly or intravenously.
[0162] In some embodiments, the antibody, or the antigen-binding fragment thereof, is administered at a dose of about 100 mg to about 5000 mg, about 300 mg to about 4500 mg, about 500 mg to about 4500 mg, about 1200 mg to about 4500 mg, about 100 mg to about 2000 mg, about 200 mg to about 1500 mg, about 300 mg to about 600 mg, about 500 mg to about 1200 mg, about 300 mg to about 1200 mg; or at a dose of about 4500 mg, a dose of about 1200 mg, a dose of about 600 mg, or a dose of about 500 mg; or as two doses of 600 mg each on the same day, or as two doses of 300 mg each on the same day.
[0163] In some embodiments, the antibody, or antigen-binding fragment thereof, is administered once, or is administered weekly.
[0164] In some embodiments, the method further comprises obtaining an epithelial lining fluid (ELF) sample from the subject.
[0165] In some embodiments, the ELF sample comprises an ELF sample from an upper airway, an lower airway, and/or an alveolar tissue.
[0166] In some embodiments, the antibody, or antigen-binding fragment thereof, reaches a concentration of about 1 μg/mL to about 100 μg/mL, about 1 μg/mL to about 80 μg/mL, about 80 μg/mL to about 100 μg/mL, about 50 μg/mL to about 100 μg/mL, about 1 μg/mL to about 50 μg/mL, about 2 μg/mL to about 25 μg/mL, or about 2 μg/mL to about 10 μg/mL, in the ELF sample of the subject.
[0167] In some embodiments, the antibody, or antigen-binding fragment thereof, has a receptor occupancy of at least 50%, 60%, 70%, 80%, or 90% in the ELF sample.
[0168] In some embodiments, the antibody, or antigen-binding fragment thereof, has a receptor occupancy of at least 50%, 60%, 70%, 80%, or 90% in the ELF sample for about 28 days after administration.
[0169] In some embodiments, the antibody, or antigen-binding fragment thereof, has a virus neutralizing titer of about 100-2000, about 200-1500, about 300-1500, or about 500-1500 in serum of the subject about 6 months after administration.
[0170] In some embodiments, the 80% virus neutralization titer (MN80) of the antibody, or antigen binding fragment thereof, at about days 0-14, about days 7-21, about 3 months, or about 6 months after administration, is about 10-6000, about 50-600, about 500-1500, about 1000-2500, about 100- 2500, about 500-2000, about 500-1500, about 400-1200, about 200-1500, about 300-1000, about 400- 800, about 400-1000, or about 500-600 in the subject.
[0171] In some embodiments, the 80% virus neutralization titer (MN80) of the antibody, or antigen binding fragment thereof, at about day 7, day 14, or day 21 after administration, is about 300-2000, about 400-600, about 600-1500, about 1100-1700, about 500-1700, about 500-1500, about 400-1200, about 400-800, about 400-1000, or about 500-600 in the serum sample of the subject.
[0172] In some embodiments, the 80% virus neutralization titer (MN80) of the antibody, or antigen binding fragment thereof, at about 3 months after administration, is about 200-1000, about 200-800, about 200-500, about 400-900, or about 400-600 in the serum sample of the subject.
[0173] In some embodiments, the 80% virus neutralization titer (MN80) of the antibody, or antigen binding fragment thereof, at about 6 months after administration, is about 10-500, about 300-500, or about 50-200 in the serum sample of the subject.
[0174] In some embodiments, the 50% virus neutralization titer (MN50) of the antibody, or antigen binding fragment thereof, at about days 0-14, about days 7-21, about 3 months, about 6 months, or about 12 months after administration, is about 100-6000, about 300-1500, about 1700-3800, about 3800-5200, about 300-5500, about 1200-4500, about 1300-4300, about 1200-4000, about 100-2500, about 500-2500, about 800-2000, about 1000-1800, about 800-1300, about 900-1100, or about 1300- 1500 in the serum sample of the subject.
[0175] In some embodiments, the 50% virus neutralization titer (MN50) of the antibody, or antigen binding fragment thereof, at about day 7, day 14 or day 21 after administration, is about 1000-4500, about 1200-4500, about 1300-4300, about 1200-3900, about 1500-4000, about 1800-3800, about 3800-4500, about 1000-1800 or about 1200-1500 in the serum sample of the subject.
[0176] In some embodiments, the 50% virus neutralization titer (MN50) of the antibody, or antigen binding fragment thereof, at about 3 months after administration, is about 800-1300, or about 900- 1100 in the serum sample of the subject.
[0177] In some embodiments, the 50% virus neutralization titer (MN50) of the antibody, or antigen binding fragment thereof, at about 6 months after administration, is about 200-500, or about 300-600 in the serum sample of the subject.
[0178] In some embodiments, the 50% virus neutralization titer (MN50) of the antibody, or antigen binding fragment thereof, at about 12 months after administration, is about 150-500, or about 200-400 in the serum sample of the subject.
[0179] In some embodiments, the virus neutralization titer is determined using a plaque reduction neutralization test (PRNT).
[0180] In some embodiments, the serum concentration of the antibody, or antigen-binding fragment thereof, is about 1-300 mg/L, about 1-250 mg/L, about 1-200 mg/L, about 1-100 mg/L, about 100-250 mg/L, about 150-200 mg/L, about 120-170 mg/L, about 1-90 mg/L, about 1-80 mg/L, about 1-70 mg/L, about 1-60 mg/L, about 1-50 mg/L, about 1-40 mg/L, about 1-30 mg/L, about 1-20 mg/L, about 1-10 mg/L, about 2-9 mg/L, about 3-8 mg/L, about 4-6 mg/L, about 5-80 mg/L, about 10-70 mg/L, or about 10-60 mg/L in the subject about 3 months after administration.
[0181] In some embodiments, the serum concentration of the antibody, or antigen-binding fragment thereof, is about 1-100 mg/L, about 1-90 mg/L, about 1-80 mg/L, about 1-70 mg/L, about 1-60 mg/L, about 1-50 mg/L, about 1-40 mg/L, about 1-30 mg/L, about 1-20 mg/L, about 1-10 mg/L, about 2-9 mg/L, about 3-8 mg/L, about 4-6 mg/L, about 5-80 mg/L, about 10-70 mg/L, about 10-60 mg/L, about 5-25 mg/L, about 10-30 mg/L, about 15-25 mg/L, about 20-100 mg/L, about 30-100 mg/L, about 50- 80 mg/L, or about 60-70 mg/L in the subject about 6 months after administration.
[0182] In some embodiments, the serum concentration of the antibody, or antigen-binding fragment thereof, is about 0.1-30 mg/L, about 1-30 mg/L, about 1-20 mg/L, about 1-10 mg/L, about 10-20 mg/L, about 0.1-3 mg/L, about 0.5-8 mg/L, about 0.5-10 mg/L, about 2-9 mg/L, about 5-15 mg/L, about 3-8 mg/L, or about 4-6 mg/L in the subject about 12 months after administration.
[0183] In some embodiments, the clearance rate for the antibody, or antigen-binding fragment thereof, is about 0.1-10 mL/d, about 0.1-5.0 mL/d, about 0.1-3.0 mL/d, about 0.5-2.5 mL/d, or about 1.0-2.0 mL/d.
[0184] In some embodiments, the steady state volume of distribution for the antibody, or antigen binding fragment thereof, is about 1-10 L, about 2-8 L, about 4-9 L, about 4-8 L, or about 5-7L.
[0185] In some embodiments, the antibody, or antigen-binding fragment thereof, is administered in combination with at least one antibody selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130 and ADI-58131.
[0186] In some embodiments, the antibody, or antigen-binding fragment thereof, is administered in combination with a vaccine.
[0187] In some embodiments, the antibody, or antigen-binding fragment thereof, is administered (a) concurrently with a COVID-19 vaccine; (b) after administration of a COVID-19 vaccine, or (c) prior to administration of a COVID-19 vaccine.
[0188] In some embodiments, administration of the antibody, or antigen-binding fragment thereof, reduces pulmonary inflammation in the subject.
[0189] In some embodiments, administration of the antibody, or antigen-binding fragment thereof, reduces the risk of COVID-19 hospitalization or death of the subject.
[0190] In some embodiments, administration of the antibody, or antigen-binding fragment thereof, reduces viral load in the subject.
[0191] In one aspect, provided herein are methods of predicting the in vivo efficacy of an anti-CoV-S antibody or antigen-binding antibody fragment in preventing or treating CoV infection.
[0192] In some embodiments, the method may comprise: (a) providing at least one first test subject and at least one second subject or a cell sample derived from at least one first test subject and at least one second subject; (b) administering the antibody or antigen-binding antibody fragment to said at least one first test subject and said at least one second subject or contacting a cell sample from said first and second subject with the antibody or antigen-binding antibody fragment; (c) infecting said at least one first test subject and said at least one second subject with CoV or pseudo CoV or a cell sample obtained from said at least one first test subject and said at least one second subject with CoV or pseudo CoV ; (d) determining whether administration of the antibody or antigen-binding antibody fragment in (b) results in one or more of the following compared to a suitable control: (I) reduction in a CoV-associated symptom; (II) reduction in the CoV viremia; (III) increase in the survival; (IV) increase in the body weight; or (V) reduced infection of cells or virus proliferation in cells of the tested cell sample compared to a control cell sample not contacted with the antibody or antigen binding antibody fragment.
[0193] In some embodiments, the method may comprise: (a) providing at least one first cell sample and at least one second cell sample; (b) contacting the at least one first cell sample with the antibody or antigen-binding antibody fragment; (c) infecting said at least one first cell sample and at least one second cell sample with CoV or pseudo CoV ; (d) determining whether the antibody or antigen binding antibody fragment results in one or more of the following compared to a suitable control: (I) increase in the cell survival; (II) reduced infection of cells; (III) reduced virus proliferation; (IV) reduced cell stress or death markers; or (V) reduced inflammatory cytokines, in cells of the tested cell sample compared to a control cell sample not contacted with the antibody or antigen-binding antibody fragment.
[0194] In some embodiments, the method may comprise: (a) providing at least one first test subject and at least one second subject or a cell sample derived from at least one first test subject and at least one second subject; (b) infecting said at least one first test subject and said at least one second subject with CoV or pseudo CoV or a cell sample derived from at least one first test subject and at least one second subject; (c) administering the antibody or antigen-binding antibody fragment to said at least one second subject or contacting a cell sample derived from at least one first test subject and at least one second subject with the antibody or antigen-binding antibody fragment; (d) determining whether administration of the antibody or antigen-binding antibody fragment in (c) results in one or more of the following: (I) reduction in a CoV-associated symptom; (II) reduction in the CoV viremia; (III) increase in the survival;(IV) increase in the body weight; or (V) reduced infection of cells or virus proliferation in cells in the tested cell sample compared to a control cell sample not contacted with the antibody or antigen-binding antibody fragment.
[0195] In some embodiments, the method may comprise: (a) providing at least one first cell sample and at least one second cell sample; (b) infecting said at least one first cell sample and at least one second cell sample with CoV or pseudo CoV ; (c) contacting the at least one first cell sample with the antibody or antigen-binding antibody fragment; (d) determining whether the antibody or antigen binding antibody fragment results in one or more of the following compared to a suitable control: (I) increase in the cell survival; (II) reduced infection of cells; (III) reduced virus proliferation; (IV) reduced cell stress or death markers; or (V) reduced inflammatory cytokines, in cells of the tested cell sample compared to a control cell sample not contacted with the antibody or antigen-binding antibody fragment.
[0196] In one aspect, provided herein are methods of screening for an antibody or antigen-binding antibody fragment that binds to a CoV or CoV-S, the method comprising whether an antibody or antigen-binding antibody fragment comprising 1, 2, 3, 4, 5, or 6 CDRs of any of the antibodies disclosed herein may comprise one or more of the following features: (i) binds to the S protein of a CoV; (ii) binds to the SI subunit of CoV-S; (iii) binds to the RBD of CoV-S; (iv) binds to the NTD of CoV-S; (v) binds to the ACE2-binding motif of CoV-S; (vi) competes with ACE2; (vii) competes with the antibody CR3022; (viii) neutralizes one or more of SARS-CoV, SARS-CoV-2, MERS-CoV, HCoV-229E, HCoV-HKUl, HCoV-NL63, or HCoV-OK43 or variants thereof; (ix) neutralizes a pseudovirus of one or more of SARS-CoV, SARS-CoV-2, MERS-CoV, HCoV-229E, HCoV-HKUl, HCoV-NL63, or HCoV-OK43 or variants thereof; (x) results in reduced infection of cells or virus proliferation in cells in a susceptible tested cell sample compared to a control cell sample not contacted with the antibody or antigen-binding antibody fragment; or (xi) prevents or treats CoV infection in vivo. During the screening, any of the antibodies disclosed herein and/or an antibody comprising one or more of the CDRs of the antibodies disclosed herein may be used as a candidate antibody or a control antibody.
[0197] In one aspect, the present disclosure also relates to compositions comprising at least one affinity-matured first anti-CoV-S antibody or antigen-binding antibody fragment and a pharmaceutically acceptable carrier or excipient.
[0198] In some embodiments, the at least one first antibody or antigen-binding antibody fragment may comprise: a VH comprising a VH CDR1, a VH CDR2, a VH CDR3; and a VL, comprising a VL CDR1 a VL CDR2, a VL CDR3, and the amino acid sequences of said VH CDR1, said VH CDR2, said VH CDR3, said VL CDR1, said VL CDR2, and said VL CDR3 are identical to the amino acid sequences of the VH CDR1, VH CDR2, VH CDR3, VL CDR1, VL CDR2, and VL CDR3, respectively, of an anti-CoV-S antibody selected from the group consisting of ADI-58120, ADI- 58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI- 58129, ADI-58130, ADI-58131.
[0199] In some embodiments, the first antibody or antigen-binding antibody fragment may comprise an Fc region, optionally wherein the Fc region may comprise an amino acid sequence of SEQ ID NOs: 411, 412, 413, 414, 415, 416, or 417.
[0200] In one embodiment, the HC and LC of the first antibody or antigen-binding antibody fragment are the HC and LC, respectively, of an anti-CoV-S antibody selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, and ADI-58131.
[0201] In certain embodiments, the composition may further comprise at least one second antibody or antigen-binding antibody fragment comprising a VH comprising a VH CDR1, a VH CDR2, a VH CDR3 and a VL, comprising a VL CDR1 a VL CDR2, a VL CDR3. In particular embodiments, the amino acid sequences of said VH CDR1, said VH CDR2, said VH CDR3, said VL CDR1, said VL CDR2, and said VL CDR3 may be identical to the amino acid sequences of the VH CDR1, VH CDR2, VH CDR3, VL CDR1, VL CDR2, and VL CDR3, respectively, of an anti-CoV-S antibody selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, and ADI-58131.
[0202] In some embodiments, the second antibody or antigen-binding antibody fragment may comprise an Lc region, optionally wherein the Lc region may comprise an amino acid sequence of SEQ ID NOs: 411, 412, 413, 414, 415, 416, or 417.
[0203] In particular embodiments, the antibody or antigen-binding antibody fragment according to the present disclosure may comprise:
(1) a HC comprising the amino acid sequence of SEQ ID NO: 10 and a LC comprising the amino acid sequence of SEQ ID NO: 210;
(2) a HC comprising the amino acid sequence of SEQ ID NO: 20 and a LC comprising the amino acid sequence of SEQ ID NO: 220;
(3) a HC comprising the amino acid sequence of SEQ ID NO: 30 and a LC comprising the amino acid sequence of SEQ ID NO: 230;
(4) a HC comprising the amino acid sequence of SEQ ID NO: 40 and a LC comprising the amino acid sequence of SEQ ID NO: 240;
(5) a HC comprising the amino acid sequence of SEQ ID NO: 50 and a LC comprising the amino acid sequence of SEQ ID NO: 250;
(6) a HC comprising the amino acid sequence of SEQ ID NO: 60 and a LC comprising the amino acid sequence of SEQ ID NO: 260;
(7) a HC comprising the amino acid sequence of SEQ ID NO: 70 and a LC comprising the amino acid sequence of SEQ ID NO: 270;
(8) a HC comprising the amino acid sequence of SEQ ID NO: 80 and a LC comprising the amino acid sequence of SEQ ID NO: 280;
(9) a HC comprising the amino acid sequence of SEQ ID NO: 90 and a LC comprising the amino acid sequence of SEQ ID NO: 290;
(10) a HC comprising the amino acid sequence of SEQ ID NO: 100 and a LC comprising the amino acid sequence of SEQ ID NO: 300;
(11) a HC comprising the amino acid sequence of SEQ ID NO: 110 and a LC comprising the amino acid sequence of SEQ ID NO: 310; or
(12) a HC comprising the amino acid sequence of SEQ ID NO: 120 and a LC comprising the amino acid sequence of SEQ ID NO: 320.
[0204] In further embodiments, the composition according to the present disclosure may comprise: (A) at least one first antibody or antigen-binding antibody fragment selected from the group consisting of the antibodies or antigen-binding antibody fragments comprising the HC and LC combination as described above; and (B) a pharmaceutically acceptable carrier or excipient.
[0205] In yet further embodiments, the composition may additionally comprise at least one second antibody or antigen-binding antibody fragment selected from the group consisting of the antibodies or antigen-binding antibody fragments comprising the HC and LC combination as described above. [0206] Additionally, the present disclosure further encompasses isolated antibodies and antigen binding antibody fragments thereof, which competes for binding with any one or more of the anti- CoV antibodies or antigen-binding antibody fragments thereof as described herein.
[0207] The present disclosure also encompasses isolated antibodies or antigen-binding antibody fragments thereof, which bind the same epitope as any one or more of the anti-CoV antibodies or antigen-binding antibody fragments thereof as described herein.
[0208] The present disclosure further encompasses affinity matured variants of any one or more of the anti-CoV antibodies or antigen-binding antibody fragments thereof as described herein.
[0209] In one aspect, disclosed herein is a method of treating a coronavirus infection by SARS-CoV, SARS-CoV-2, and/or another coronavirus optionally selected from the group consisting of MERS- CoV, HCoV-HKUl, HCoV-OC43, HCoV-229E, and HCoV-NL63 in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of an antibody, or antigen-binding antibody fragment thereof, which binds the same epitope as ADI-58125, and/or which competes for binding with ADI-58125.
[0210] In one aspect, disclosed herein is a method of decreasing the risk of mortality, hospitalization, mechanical ventilation, or a combination thereof in a patient infected by SARS-CoV, ARS-CoV-2, and/or another coronavirus optionally selected from the group consisting of MERS-CoV, HCoV- HKU1, HCoV-OC43, HCoV-229E, and HCoV-NL63, the method comprising administering to the subject a therapeutically effective amount of an isolated antibody, or antigen-binding antibody fragment thereof, which binds the same epitope as ADI-58125, and/or which competes for binding with ADI-58125.
[0211] In another aspect, disclosed herein is a method of preventing infection of a subject by SARS- CoV, SARS-CoV-2, and/or another coronavirus optionally selected from the group consisting of MERS-CoV, HCoV-HKUl, HCoV-OC43, HCoV-229E, and HCoV-NL63, the method comprising administering to the subject a therapeutically effective amount of an isolated antibody, or antigen binding antibody fragment thereof, which binds the same epitope as ADI-58125, and/or which competes for binding with ADI-58125.
[0212] In another aspect, the present invention also provides a method for treating a coronavirus infection in a subject in need thereof, the method comprising administering to the subject an isolated antibody, or antigen-binding fragment thereof, wherein the antibody, or antigen-binding fragment thereof, comprises a heavy chain variable region (VH) comprising a VH CDR1 comprising SEQ ID NO:52, a VH CDR2 comprising SEQ ID NO:54, and a VH CDR3 comprising SEQ ID NO:56, and a light chain variable region (VL) comprising a VL CDR1 comprising SEQ ID NO:252, a VL CDR2 comprising SEQ ID NO:254, and a VL CDR3 comprising SEQ ID NO:256; and wherein the antibody, or antigen-binding fragment thereof, is administered at a dosage of at least 300 mg within 5, 4, 3, 2, or 1 days of symptom onset.
[0213] In some embodiments, the antibody, or antigen-binding fragment thereof, is administered at a dosage of about 300 mg. In some embodiments, the antibody, or antigen-binding fragment thereof, is administered at a dosage of about 500 mg. In some embodiments, the antibody, or antigen-binding fragment thereof, is administered at a dosage of about 600 mg. In some embodiments, the antibody, or antigen-binding fragment thereof, is administered at a dosage of about 1200 mg. In some embodiments, the antibody, or antigen-binding fragment thereof, is administered at a dosage of about 4500 mg.
[0214] In some embodiments, the subject has at least one risk factor which renders them more prone to a poor clinical outcome. In some embodiments, the at least one risk factor is selected from the group consisting of: an old age selected from the group consisting of over 55, over 60 or over 65 years old; diabetes, a chronic respiratory condition, obesity, hypertension, a cardiac or cardiovascular condition, a chronic inflammatory or autoimmune condition, and an immune compromised status. In some embodiments, the subject is immunocompromised.
[0215] In some embodiments, the subject is at a high risk of disease progression. In some embodiments, the subject is (a) age 50 years or above with no comorbid conditions or additional risk factors for progression of COVID-19; (b) between about 18 to about 50 years old and with one or more preexisting medical conditions selected from the group consisting of obesity, diabetes, chronic kidney disease, chronic lung disease, cardiac disease, sickle cell disease or thalassemia, solid organ or blood stem cell transplant recipient, other immunodeficiency due to underlying illness or immunosuppressant medication, Down Syndrome, stroke or cerebrovascular disease, substance use disorder, and pregnancy; or (c) between about 12 to about 17 years old and with one or more
preexisting medical conditions selected from the group consisting of obesity, diabetes, chronic kidney disease, sickle cell disease or thalassemia, congenital or acquired heart disease, neurodevelopmental disorder, a medically-related technological dependence, asthma or chornic respiratory disease, solid organ or blood stem cell transplant recipient, other immunodeficiency due to underlying illness or immunosuppressant medication, Down Syndrome, stroke or cerebrovascular disease, substance use disorder, and pregnancy. In some embodiments, the subject is age 50 or above with no comorbid conditions or additional risk factors for progression of COVID-19. In some embodiments, the subject has hypertension with at least one medication prescribed or recommended. In some embodiments, the subject has moderate to severe asthma requiring daily therapy.
[0216] In another aspect, the present invention provides a composition for use in inducing an immune response against CoV in a subject at high risk of COVID disease progression, wherein the composition comprises an antibody, or antigen-binding fragment thereof, comprises a heavy chain variable region (VH) comprising a VH CDR1 comprising SEQ ID NO:52, a VH CDR2 comprising SEQ ID NO:54, and a VH CDR3 comprising SEQ ID NO:56, and a light chain variable region (VL) comprising a VL CDR1 comprising SEQ ID NO:252, a VL CDR2 comprising SEQ ID NO:254, and a VL CDR3 comprising SEQ ID NO:256; and wherein the antibody, or antigen-binding fragment thereof, is administered within 5, 4 ,3, 2, or 1 days of symptom onset.
[0217] In another aspect, the present invention provides a composition for use in treating or preventing a coronavirus infection in a subject at high risk of COVID disease progression, wherein the composition comprises an antibody, or antigen-binding fragment thereof, comprises a heavy chain variable region (VH) comprising a VH CDR1 comprising SEQ ID NO:52, a VH CDR2 comprising SEQ ID NO:54, and a VH CDR3 comprising SEQ ID NO:56, and a light chain variable region (VL) comprising a VL CDR1 comprising SEQ ID NO:252, a VL CDR2 comprising SEQ ID NO:254, and a VL CDR3 comprising SEQ ID NO:256; and wherein the antibody, or antigen-binding fragment thereof, is administered within 5, 4, 3, 2, or 1 days of symptom onset.
[0218] In another aspect, the present invention provides a composition for use in treating a symptom of a coronavirus infection in a subject at high risk of COVID disease progression, wherein the composition comprises an antibody, or antigen-binding fragment thereof, comprises a heavy chain variable region (VH) comprising a VH CDR1 comprising SEQ ID NO:52, a VH CDR2 comprising SEQ ID NO:54, and a VH CDR3 comprising SEQ ID NO:56, and a light chain variable region (VL) comprising a VL CDR1 comprising SEQ ID NO:252, a VL CDR2 comprising SEQ ID NO:254, and a VL CDR3 comprising SEQ ID NO:256; and wherein the antibody, or antigen-binding fragment thereof, is administered within 5, 4, 3, 2, or 1 days of symptom onset.
[0219] In another aspect, the present invention provides a composition for use in decreasing the risk of mortality, hospitalization, mechanical ventilation, or a combination thereof, in a subject at high risk of COVID disease progression, wherein the composition comprises an antibody, or antigen-binding
fragment thereof, comprises a heavy chain variable region (VH) comprising a VH CDR1 comprising SEQ ID NO:52, a VH CDR2 comprising SEQ ID NO:54, and a VH CDR3 comprising SEQ ID NO:56, and a light chain variable region (VL) comprising a VL CDR1 comprising SEQ ID NO:252, a VL CDR2 comprising SEQ ID NO:254, and a VL CDR3 comprising SEQ ID NO:256; and wherein the antibody, or antigen-binding fragment thereof, is administered within 5, 4, 3, 2, or 1 days of symptom onset.
BRIEF DESCRIPTION OF THE DRAWINGS [0220] The patent or application file contains at least one drawing executed in color. Copies of this patent or patent application publication with color drawing(s) will be provided by the Office upon request and payment of the necessary fee.
[0221] Figure 1 depicts the mean serum concentration of ADI-58125 in 8 subjects following a single IM adminsitration of 300 mg ADI-58125 over a period of 21 days.
[0222] Figure 2 depicts the quantitative systems pharmacology/physiologically based pharmacokinetic (QSP/PBPK) model-predicted and observed ADI-58125 concentrations following a 300 mg IM dose across the 21 -day sampling interval. The line represents the mediam model -predicted ADI-58125 profile, shaded area represents the 90% prediction interval, and black circles represent observed ADI-58125 concentrations.
[0223] Figure 3 depicts the serum neutralizing titers from participants receiving a single 300 mg IM dose of ADI-58125.
[0224] Figure 4 is a schematic depicting the experimental procedure for a plaque reduction neutralization test (PRNT)
[0225] Figure 5 is a schematic depicting the experimental procedure for a post-infection assay to measure the concentration of antibody required to prevent viral replication in infected cells.
[0226] Figure 6 depicts the concentration curves of ADI-58125 for preventing replication of SARS- CoV-2 D614G and beta B.1.351 variants in infected cells.
[0227] Figure 7 depicts the neutralizing activity of ADI-58125 and ADI-58122 against authentic SARS-CoV-2 variants including the Alpha/B.1.1.7, the Beta/B.1.351, the Gamma/P.1, the Delta/B.1.617, and Omicron variants. Additional antibodies were also shown as comparators.
[0228] Figure 8A depicts the neutralizing activity of ADI-58125 and ADI-58122 against pseudoviruses encoding spike proteins of circulating SARS-CoV-2 variants. Figure 8B depicts the neutralizing activity of ADI-58125 and ADI-58122 against pseudoviruses encoding SARS-CoV-2 spike proteins with sing!e-/doubIe-spike mutations, in a pseudovirus assay. The neutralizing activity was expressed as a fold change relative to the D614G reference variant. Figure 8C depicts the neutralizing activity of ADI-58125 against SARS-CoV-2 variants associated with resistance to EUA mAbs. The points represent neutralizing activity of ADI-58125 against SARS-CoV-2 variants.
[0229] Figures 9A, 9B, 9C, and 9D depict an overview of QSP/PBPK model. Figure 9A depicts a tissue-level diagram. Figure 9B depicts a cellular-level diagram specifically for the upper airway, lower airway, and alveolar spaces within the lung. Figure 9C depicts the predicted median ADI- 58125 concentration in NHP serum following a single ADI-58125 10 mg/kg IM dose with observed NHP ADI-58125 concentration data overlaid (black dots). The shaded area represents the 90% prediction interval. Figure 9D depicts the predicted median human serum and ELF PK in humans following a single ADI-58125 300 mg IM dose with observed human serum (red line) ADI-58125 concentration data overlaid (black dots), ELF upper airway (blue line), and ELF lower airway (gold). The shaded area represents the 90% prediction interval. CLup, rate of pinocytosis of antibody entry and exit from the endosomal space; CLup-epi, rate of pinocytosis of antibody entry and exit from the epithelial space; FR, fraction of FcRn bound antibody that recycles to the vascular space; L, lymphatic flow rate; LG, large, kde , degradation constant; k0ff, first-order dissociation rate constant of antibody from FcRn; kon, second-order association rate constant for binding of antibody to FcRn; Q, blood or tissue flow rate; SM, small.
[0230] Figures 10A-10B depict a QSP/PBPK model forecast of ADI-58125 300 mg IM in adults. Figure 10A depicts the QSP/PBPK predicted median ADI-58125 concentration following a single 300 mg IM injection in different compartments overlaid by the threashold associated with 95% viral growth suppression (as indicated by the dotted line) for the SARS-CoV-2 Beta variant in a post infection assay. The shaded area represents the 90% prediction interval. Figure 10B depicts the receptor occupancy (RO) of ADI-58125 at various Delta variant virion densities (107, 10s and 109) after a single 300 mg IM injection expressed as percent occupancy with the dotted line representing the threshold for 90% RO.
[0231] Figure 11 is a schematic representation for the in vivo efficacy study in hamsters. Hamsters were treated with a range of ADI-58125 doses (9.25 - 2000 μg) or control mAh 24 hours prior to challenge with SARS-2/WA-1 to evaluate prophylactic efficacy of ADI-58125.
[0232] Figures 12A-12B depict the impact of ADI-58125 on weight gain and lung pathology in SARS-CoV-2 -infected Syrian hamsters. Specifically, Figure 12A depicts the changes in weight from baseline on Day 6. Figure 12B depicts the total lung pathology score. Bars represent mean ± standard deviation. Dotted line represents no change in weight from baseline. Statistical comparison was calculated using unpaired, two-sided t-tests (A) and unpaired two-sided Mann-Whiteney tests (B): *P<0.05; **** P<0.0001 vs controls.
[0233] Figure 13 depicts the impact of ADI-58125 on viral load in SARS-CoV-2 -infected Syrian hamsters. Bars represent mean ± standard deviation. Dotted line represents limit of detection. Statistical comparison was calculated using unpaired, two-sided t-tests: **P<0.005; *** P<0.001; ****p<0.0001 vs controls.
[0234] Figure 14 is a schematic representation for the in vivo efficacy study in non-human primates (NHP). Rhesus macaques were treated with ADI-58125 at 5 mg/kg or 25 mg/kg, or control mAh (25 mg/kg) 3 days prior to challenge with SARS-2/WA-1 to evaluate prophylactic efficacy of ADI-58125. [0235] Figure 15 depicts the impact of ADI-58125 on viral replication in Rhesus macaques, specifically, the impact on replication of sub-genomic viral RNA in bronchoalveolar lavage (BAL), nasopharyngeal (NP) and oropharyngeal (OP) samples.
[0236] Figure 16 depicts the impact of ADI-58125 on viral titre in Rhesus macaques.
[0237] Figure 17 depicts the mean serum concentration of ADI-58125 in 8 subjects following a single IM adminsitration of 300 mg ADI-58125 over a period of 3 months.
[0238] Figure 18 depicts the serum neutralizing titers from participants receiving a single 300 mg IM dose of ADI-58125 as compared to peak responses following AZD1222 and mRNA-1273 vaccination. Statistical analysis is by 2-tailed Mann-Whitney U test (*P<0.05; **P<0.01; ***P<0.0001).
[0239] Figure 19 depicts the mean serum concentration of ADI-58125 in subjects following a single IM adminsitration of 300 mg ADI-58125, a single IM adminsitration of 600 mg ADI-58125, and a single IV administration of 500 mg ADI-58125 over a period of 6 months.
[0240] Figure 20 depicts the relationship between ADI-58125 serum concentration and sVNA over a period of 6 months. The line represents the mean linear regression. The hading represents 95% confidence interval. Black circles represent individual time matched ADI-58125 serum concentration and MN50 sVNA titer measurements. Linear regression excluded samples that were taken following SARS-CoV-2 vaccination from participants who received vaccination during the trial.
[0241] Figures 21A, 21B, and 21C depict the simulated human body weight (A) and distribution of estimated KD,FcRn values for other extended half-life mAbs in healthy human adults (log-normal, B) and NHPs (normal, C).
[0242] Figures 22A-22B depict the observed (dots) and model-forecasted NHP median (90% prediction interval) serum ADI-58125 PK profiles based on distribution of NHP KD,FcRn values for other extended half-life mAbs following intravenous (IV; A) and intramuscular (IM; B) administration. Dots represent the raw observed data. Solid lines represent simulated medians. The shaded area represents the 90% prediction interval.
[0243] Figures 23A-23B depict the observed (dots) and optimized QSP model-predicted NHP median (90% prediction interval) serum ADI-58125 concentration-time profiles in NHPs following intravenous (IV; A) and intramuscular (IM; B) administration. Dots represent the raw observed data. Solid lines represent simulated medians. The shaded area represents the 90% prediction interval. [0244] Figures 24A-24D depict the observed (dots) versus QSP model-predicted median (90% prediction interval) serum ADI-58125 PK profiles in healthy adult participants predicted a priori based on the distribution of human KD,FcRn values for other extended half-life mAbs (A, C) and after
optimization (B, D). Dots represent the raw observed data. Solid lines represent simulated medians. The shaded area represents the 90% prediction interval.
[0245] Figures 25A-25C depict the QSP model-predicted median (90% prediction interval) serum ADI-58125 PK profiles following a single IM 150 mg (A), 300 mg (B), and 450 mg (C) injection in humans predicted a priori based on the distribution of human KD,FcRn values for other extended half- life mAbs (as shown in Figures 21 A-B). Dashed line represents 100 * in vitro IC90 of 0.011 μg/mL or 1.1 mg/L against the USA-WA1/2020 variant.
[0246] Figures 26A-26C depict the optimized QSP model-predicted median (90% prediction interval) serum ADI-58125 PK profiles following a single IM 150 mg (A), 300 mg (B), and 450 mg (C) injection in humans. Dashed line represents 100 * in vitro IC90 of 0.011 μg/mL or 1.1 mg/L against the USA-WA1/2020 variant.
[0247] Figure 27 depicts the serum neutralizing titers (geometric mean MN80) from participants receiving a single 300 mg IM dose of ADI-58125 as compared to peak titers with 2-dose mRNA-1273 vaccination. “Statistical comparisons for Delta were based on prelilminary results. bPeak vaccine response timepoint: 7 to 31 days post 2nd dose; cExeludes samples taken following SARS-CoV-2 vaccination. Statistical comparisons of titres achieved by ADI-58125 against each variant were performed using 2-sided Mann Whitney U tests: *P<0.05; **P<0.01; ***P<0.001; ns, non-significant vs mRNA-1273 vaccination response against the corresponding variant.
[0248] Figure 28 depicts the visual predictive check plots for the base ADI-58125 pharmacokinetic model.
[0249] Figure 29A depicts the key binding residues in SARS-CoV-2 S protein for ADI-58125, and the strong conservation of these residues among global circulating SARS-CoV-2 variants.
Frequencies were calculated from 3,928,364 sequences deposited in GISAID (Global Initiative on Sharing All influenza Data) as of Oct 15, 2021. Figure 29B depicts the key binding residues in SARS-CoV-2 S protein for class 1 (e.g., LY-C0VOI6, REGN10933), class 2 (e.g., P2B-2F6, LY- CoV555, COV2-2196) and class 3 (e.g., REGN10987, COV2-2130, LY-CoV1404) antibodies.
[0250] Figure 30 depicts that the newly emerged Omicron (B.1.1.529) variant contains mutations in the epitopes of all three public classes of antibodies.
[0251] Figure 31 depicts that ADI-58125 binds to an epitope distinct from those recognized by common elicited nAbs.
[0252] Figure 32A depicts the non-overlapping epitopes between ADI-58125 and other clinical stage/EUA authorized antibodies. Figure 32B depicts the non-overlapping epitopes between ADI- 58122 and ADI-58125.
[0253] Figure 33 depicts the locally extensive area of red discoloration and consolidation (black arrows) in the hilar region of the right caudal lung lobe at 6 dpi. RM 1221.
[0254] Figure 34 depicts the cut section of right caudal lung lobe showing the locally extensive region of consolidation/non-inflating (black arrow) at 6 dpi. RM 1221.
[0255] Figures 35A-35E depict the right caudal lung lobe, central section. Figure 35A. Multifocal moderate interstitial pneumonia at 6 dpi, 2x, H&E, RM 1221. Figure 35B, boxed area from Figure 35A, Moderate inflammation expanding the alveolar septa and extending into the alveolar lumina and moderate type II pneumocyte hyperplasia at 6 dpi, lOx, H&E, RM 1221. Figure 35C, boxed area from Figure 35B, alveolar septa were expanded by moderate numbers of lymphocytes, histiocytes, neutrophils and fibrin, which extended into lumen. Multifocally alveolar septa were lined by type II pneumocytes (arrows) at 6 dpi, 20x, H&E, RM 1221 Figure 35D. Rare IHC positive labeling in areas of inflammation, 2x, nucleocapsid protein for SARS-CoV-2, RM 1221. Figure 35E, boxed areas from Figure 35D, rare IHC positive labeling in areas of inflammation, 20x, nucleocapsid protein for SARS- CoV-2, RM 1221.
[0256] Figures 36A-36B depict nasal turbinate. Figure 36A. Locally extensive region of ulceration with subadjacent inflammation, hemorrhage and edema at 5 dpi, 4x, H&E, RM 7137. Figure 36B, boxed area from Figure 36A: Mucosal ulceration with mild to moderate lymphoplasmacytic and neutrophilic inflammation with necrosis, hemorrhage and edema at 5 dpi, lOx, H&E, RM 7137.
[0257] Figures 37A-37D depict right caudal lung lobe, central section. Figure 37A. Multifocal mild inflammation surrounding blood vessels, expanding alveolar septa and occasionally extending into the lumina at 5 dpi, 2x, H&E, RM 7175. Figure 37B, boxed area from Figure 37A: Mild inflammation composed predominately of histiocytes and lymphocytes with fewer neutrophils, around vessels and expanding septa at 5 dpi, lOx, H&E, RM 7175. Figure 37C. IHC labeling was not identified in areas of inflammation at 5 dpi, 2x, nucleocapsid protein for SARS-CoV-2, RM 7175. Figure 37D. IHC labeling was not identified in areas of inflammation at 5 dpi, lOx, nucleocapsid protein for SARS- CoV-2, RM 7175.
[0258] Figures 38A-38D depict lung, left caudal lobe, central section. RM 1430. Figure 38A. Multifocal mild inflammation surrounding blood vessels, expanding alveolar septa and extending into the alveolar lumina at 6 dpi, 2x, H&E, RM 1430. Figure 38B, boxed area from Figure 38A: Mild inflammation composed predominately of histiocytes and lymphocytes with fewer neutrophils, around vessels and expanding septa and within the alveolar lumina at 6 dpi, lOx, H&E, RM 1430. Figure 38C. IHC labeling was not identified in areas of inflammation at 6 dpi, 2x, nucleocapsid protein for SARS-CoV-2, RM 1430. Figure 38D. IHC labeling was not identified in areas of inflammation at 6 dpi, lOx, nucleocapsid protein for SARS-CoV-2, RM 1430.
[0259] Figure 39 depicts mild red discoloration for all lung lobes at 5 dpi. RM 1294.
[0260] Figure 40 depicts multifocal mild red discoloration throughout all lobes with locally extensive fibrinous adhesions noted from the left caudal lung lobe to the thoracic cavity at 5 dpi. RM 5240.
[0261] Figure 41 depicts average fold change in IC50 for ADI-58125 relative to the D614G reference strain in a lentiviral pseudovirus assay.
[0262] Figure 42A shows the neutralization curves for ADI-58125 and ADI-58122 against Omicron BA.l variant (red) and the D614G reference strain (black) using a pseudo virus neutralization assay. [0263] Figue 42B shows the neutralization curves for ADI-58125 against D614G and Omicron sublineages (BA.l, BA.1.1 and BA.2) using a pseudovirus neutralization assay.
[0264] Figure 43 shows the neutralization curves for ADI-58125, ADI-58122, ADI-58127 and other commercial antibodies against Victoria and Omicron sublineages (BA.l, BA.1.1 and BA.2) in an authentic neutralization assay.
[0265] Figure 44 shows the neutralization data for ADI-58125, ADI-58122, ADI-58127 and other commercial antibodies against Victoria and all variants of concern, Alpha, Beta, Gamma, Delta and Omicron (BA.l., BA.1.1, BA.2) in an authentic neutralization assay.
[0266] Figure 45 depicts the correlation between serum neutralizing antibody titers induced by vaccination or mAh administration and reported vaccine or mAh efficacy against symptomatic COVID-19. Geometric mean serum neutralizing titers for samples obtained from AZD1222 and BNT162b2 vaccinees at the indicated timepoints against the indicated variants are plotted against reported vaccine efficacy at matched timepoints against the indicated variants. Red and black dotted lines indicate predicted month 3 serum neutralizing antibody titers against the Omicron variant following administration of 300 mg IM AZD7442 or 1200 mg IM ADI-58125, respectively. Predicted serum neutralizing antibody titers were calculated by dividing the estimated serum concentration of mAb(s) at the indicated time point by the mAh neutralization IC50(s) against Omicron. *Protective efficacy data shown for REGEN-COV (1200 mg SC) and AZD7442 (300 mg IM) at 8 and 6.5 months, respectively, was obtained from published reports (AstraZeneca 2021; O'Brien 2021; Regeneron 2021). Predicted serum neutralizing antibody titers shown for AZD7442 and REGEN- COV against the Delta variant were calculated by dividing the serum antibody concentration estimated from published PK data at the indicated timepoint by the mAh neutralization IC50(s) against Delta. Abbreviations: FRNT50=50% focus reduction neutralization test; 3M=month 3.
[0267] Figure 46 depicts the predicted median (10%-90% PI) ADI-58125 serum concentration-time profile following single 1200 mg IM dose. Simulation from preliminary PopPK model developed using data from the first-in-human study. Solid black line and shaded area represent median and 80% (10%-90%) prediction interval, respectively. Solid red line indicates the median serum concentration of ADI-58125 associated with the target threshold serum neutralizing titer of 1:100. Blue and green dashed/dotted lines represent the median IC90 (dashed) and IC50 (dotted) values for ADI-58125 against the Omicron variant in the ELF assuming an ELF: serum ratio of 10% (blue) or 15% (green). Abbreviations: ELF=epithelial lining fluid; sVNT=serum virus neutralizing titer.
[0268] Figure 47 depicts the predicted median (10%-90% PI) ADI-58125 serum concentration-time profile following single 1200 mg IV dose. Simulation from preliminary PopPK model developed using data from the first-in-human study. Solid black line and shaded area represent median and 80% (10%-90%) prediction interval, respectively. Solid red line indicates the median serum concentration of ADI-58125 associated with the target threshold serum neutralizing titer of 1:100. Blue and green dashed/dotted lines represent the median IC90 (dashed) and IC50 (dotted) values for ADI-58125 against the Omicron variant in the ELF assuming an ELF: serum ratio of 10% (blue) or 15% (green). Abbreviations: ELF=epithelial lining fluid; sVNT=serum virus neutralizing titer.
[0269] Figure 48 depicts the neutralization curves of authentic Omicron virus by ADI-58125 and EUA mAbs. Antibody neutralization of authentic Omicron virus (B.1.1.529), as determined by a focus reduction neutralization assay. Data points and error bars represent the mean and standard deviation. Dotted lines indicate 50% and 90% neutralization.
[0270] Figure 49 depicts the goodness-of-fit plots for the ADI-58125 PK model.
[0271] Figure 50 depicts the key ADI-58125 PK parameter distributions across all doses (300mg IM, 500 mg IV, and 600 mg IM). CL: clearance. T - half-life. Vss: steady state volume of distribution.
[0272] Figure 51 depicts the population median predicted concentrations of ADI-58125 over time with IV and IM administration.
[0273] Figure 52A provides a graph comparing binding affinity of ADI-58125 and affinity maturation progenies for the SI from the Omicron BA.l varaint. Figure 52B provides a graph comparing the ability of ADI-58125 and affinity maturation progenies to neutralize the Omicron BA.l varaint.
[0274] Figure 53A provides a graph comparing binding affinity of affinity maturation ADI-58125 progenies for different variants of concern. Figure 53B provides a graph comparing the ability of affinity maturation ADI-58125 progenies to neutralize different variants of concern.
[0275] Figure 54 depicts the median (90% confidenence interval) ADI-58125 concentration-time profile following a 300 mg IM dose for phase 1 and phase 2/3 prevention and treatment studies. Shaded areas represent 90% CL Solid line represents median.
[0276] Figure 55 depicts the correlation between serum neutralizing antibody titers induced by vaccination or mAh administration and reported vaccine or mAh efficacy against symptomatic COVID-19. Geometric mean serum neutralizing titers for samples obtained from AZD1222 and BNT162b2 vaccinees at the indicated timepoints against the indicated variants are plotted against reported vaccine efficacy at matched timepoints against the indicated variants. Serum concentrations from population PK model based on Phase 2/3 data of ADI-58125 on days 55 and 76 (Omicron BA.1/BA1.1), and 90 (Delta) were used to calculate predictive nAb titers and corresponding efficacy for the Delta and Omicron variants and then compared to clinical efficacy results from the EVADE
clinical trial in prevention of COVID-19. Abbreviations: FRNT50=50% focus reduction neutralization test.
DETAILED DESCRIPTION
A. Definitions
[0277] It is to be understood that this disclosure is not limited to the particular methodology, protocols, cell lines, animal species or genera, and reagents described, as such may vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only and is not intended to limit the scope of the present disclosure, which will be limited only by the appended claims. As used herein the singular forms "a", "and", and "the" include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to "a cell" includes a plurality of such cells and reference to "the protein" includes reference to one or more proteins and equivalents thereof known to those skilled in the art, and so forth. All technical and scientific terms used herein have the same meaning as commonly understood to one of ordinary skill in the art to which this disclosure belongs unless clearly indicated otherwise.
[0278] Spike protein (S protein ): As used herein, unless stated otherwise S protein includes any coronavirus form of S protein. The term coronavirus S protein (“CoV-S”) is used to describe the S protein of any coronaviruses. In particular, the “SARS-CoV-S” and “SARS-CoV-2-S” encompass the S protein of SARS-CoV and of SARS-CoV-2. SEQ ID NO: 401 is an exemplary polypeptide sequence of SARS-CoV-S, comprising 1288 amino acids (Accession# PDB: 6VSB_B). SEQ ID NO: 403 is an exemplary polypeptide sequence of SARS-CoV-2-S, comprising 1273 amino acids (GenBank: QHD43416.1). SEQ ID NO: 402 (3864 nucleotides) encodes the SARS-CoV-S (SEQ ID NO: 401) and SEQ ID NO: 404 (3822 nucleotides, NC_045512:21563.25384, also see the corresponding region of GenBank: MN908947) encodes SARS-CoV -2-S (SEQ ID NO: 403).
[0279] In some embodiments, the “SARS-CoV-S” and “SARS-CoV -2-S” encompass any mutants, splice variants, isoforms, orthologs, homologs, and variants of SEQ ID Nos 401 and 403. In some embodiments, the CoV-S comprises a polypeptide sequence having at least 85%, 90%, 95%, 96%, 97%, 98%, or 99% identity to either SEQ ID NO: 401 or SEQ ID NO: 403.
[0280] “Effective treatment or prevention of CoV infection” herein refers to eliminating CoV from the subject or preventing the expansion of CoV in the subject or eliminating or reducing the symptoms such as fever, cough, shortness of breath, runny nose, congestion, conjunctivitis, and/or gastrointestinal symptoms after administration of an effective amount of an anti-CoV-S antibody or antigen-binding fragment thereof. In some instances, effective treatment may eliminate the need for the subject to be placed on a ventilator or reduce the time the subject needs to be on a ventilator. The treatment may be effected as a monotherapy or in association with another active agent such as an antiviral agent or anti-inflammatory agent by way of example.
[0281] As used herein, "treatment" is an approach for obtaining beneficial or desired clinical results. For purposes of this disclosure, beneficial or desired clinical results include, but are not limited to, one or more of the following: improvement in any aspect of COV-S-related conditions such as fever or cough. For example, in the context of CoV infection treatment this includes lessening severity, alleviation of fever, cough, shortness of breath, and other associated symptoms, reducing frequency of recurrence, increasing the quality of life of those suffering from the CoV-related symptoms, and decreasing dose of other medications required to treat the CoV-related symptoms. Other associated symptoms include, but are not limited to, diarrhea, conjunctivitis, loss of smell, and loss of taste. Still other symptoms which may be alleviated or prevented include inflammation, cytokine storm and/or sepsis.
[0282] "Reducing incidence" or “prophylaxis” or “prevention” means any of reducing severity for a particular disease, condition, symptom, or disorder (the terms disease, condition, and disorder are used interchangeably throughout the application). Reduction in severity includes reducing drugs and/or therapies generally used for the condition by, for example, reducing the need for, amount of, and/or exposure to drugs or therapies. Reduction in severity also includes reducing the duration, and/or frequency of the particular condition, symptom, or disorder (including, for example, delaying or increasing time to next episodic attack in an individual). This further includes eliminating the need for the subject to be placed on a ventilator or reducing the time the subject needs to be on a ventilator. [0283] "Ameliorating" one or more symptoms of CoV infection-related conditions means a lessening or improvement of one or more symptoms of the condition, e.g., fever or cough or shortness of breath as compared to not administering an anti-CoV-S antagonist antibody. "Ameliorating" also includes shortening or reduction in duration of a symptom. Again, this may include eliminating the need for the subject to be placed on a ventilator or reducing the time the subject needs to be on a ventilator. [0284] As used herein, "controlling CoV-related symptom” or “controlling” another CoV-S-related condition refers to maintaining or reducing severity or duration of one or more symptoms of the condition (as compared to the level before treatment). For example, the duration or severity or frequency of symptoms is reduced by at least about any of 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100% in the individual as compared to the level before treatment. The reduction in the duration or severity, or frequency of symptoms can last for any length of time, e.g., 2 weeks, 4 weeks (1 month), 8 weeks (2 months), 16 weeks (3 months), 4 months, 5 months, 6 months, 9 months, 12 months, etc.
[0285] As used therein, "delaying" the development of a CoV-S-related condition such as shortness of breath, bronchitis, or pneumonia e.g., interstitial), means to defer, hinder, slow, retard, stabilize, and/or postpone progression of the condition or disease. This delay can be of varying lengths of time, depending on the history of the condition or disease and/or individuals being treated. As is evident to one skilled in the art, a sufficient or significant delay can, in effect, encompass prevention, in that the
individual does not develop symptoms. A method that "delays" development of the symptom is a method that reduces probability of developing the symptom in a given time frame and/or reduces extent of the symptoms in a given time frame, when compared to not using the method. Such comparisons are typically based on clinical studies, using a statistically significant number of subjects. [0286] "Development" or "progression" of a CoV-related condition such as cough or fever means initial manifestations and/or ensuing progression of the disorder. Development of cough or fever can be detectable and assessed using standard clinical techniques as well known in the art. However, development also refers to progression that may be undetectable. For purpose of this disclosure, development, or progression refers to the biological course of the symptoms. "Development" includes occurrence, recurrence, and onset. As used herein "onset" or "occurrence" of a condition includes initial onset and/or recurrence.
[0287] As used herein, an "effective dosage" or "effective amount" of drug, compound, or pharmaceutical composition is an amount sufficient to effect beneficial or desired results. For prophylactic use, beneficial or desired results include results such as eliminating or reducing the risk, lessening the severity, or delaying the outset of the disease, including biochemical, histological, and/or behavioral symptoms of the disease, its complications and intermediate pathological phenotypes presenting during development of the disease. For therapeutic use, beneficial or desired results include clinical results such as reducing symptom intensity, duration, or frequency, and decreasing one or more symptoms resulting from CoV infection, including its complications and intermediate pathological phenotypes presenting during development of the disease, increasing the quality of life of those suffering from the disease, decreasing the dose of other medications required to treat the disease, enhancing effect of another medication, and/or delaying the progression of the disease of patients, eliminating the need for the subject to be placed on a ventilator or reducing the time the subject needs to be on a ventilator.
[0288] An effective dosage can be administered in one or more administrations. For purposes of this disclosure, an effective dosage of drug, compound, or pharmaceutical composition is an amount sufficient to accomplish prophylactic or therapeutic treatment either directly or indirectly. As is understood in the clinical context, an effective dosage of a drug, compound, or pharmaceutical composition may or may not be achieved in conjunction with another drug, compound, or pharmaceutical composition. Thus, an "effective dosage" may be considered in the context of administering one or more therapeutic agents, and a single agent may be considered to be given in an effective amount if, in conjunction with one or more other agents, a desirable result may be or is achieved.
[0289] A “suitable host cell” or “host cell” generally includes any cell wherein the subject anti-CoV- S antibodies and antigen-binding fragments thereof can be produced recombinantly using techniques and materials readily available. For example, the anti-CoV-S antibodies and antigen-binding
fragments thereof of the present disclosure can be produced in genetically engineered host cells according to conventional techniques. Suitable host cells are those cell types that can be transformed or transfected with exogenous DNA and grown in culture, and include bacteria, fungal cells (e.g., yeast), and cultured higher eukaryotic cells (including cultured cells of multicellular organisms), particularly cultured mammalian cells, e.g., human or non-human mammalian cells. In an exemplary embodiment these antibodies may be expressed in CHO cells. Techniques for manipulating cloned DNA molecules and introducing exogenous DNA into a variety of host cells are disclosed by Sambrook et ai, Molecular Cloning: A Laboratory Manual, 2nd ed., Cold Spring Harbor, N.Y.: Cold Spring Harbor Laboratory Press (1989), and Current Protocols in Molecular Biology, Ausubel et ai, editors, New York, NY: Green and Wiley and Sons (1993).
[0290] In some exemplary embodiments the antibodies may be expressed in mating competent yeast, e.g., any haploid, diploid or tetraploid yeast that can be grown in culture. Yeast useful in fermentation expression methods may exist in a haploid, diploid, or other polyploid form.
[0291] A “selectable marker” herein refers to a gene or gene fragment that confers a growth phenotype (physical growth characteristic) on a cell receiving that gene as, for example through a transformation event. The selectable marker allows that cell to survive and grow in a selective growth medium under conditions in which cells that do not receive that selectable marker gene cannot grow. Selectable marker genes generally fall into several types, including positive selectable marker genes such as a gene that confers on a cell resistance to an antibiotic or other drug, temperature when two temperature sensitive (“ts”) mutants are crossed or a ts mutant is transformed; negative selectable marker genes such as a biosynthetic gene that confers on a cell the ability to grow in a medium without a specific nutrient needed by all cells that do not have that biosynthetic gene, or a mutagenized biosynthetic gene that confers on a cell inability to grow by cells that do not have the wild type gene; and the like.
[0292] An “expression vector” herein refers to DNA vectors containing elements that facilitate manipulation for the expression of a foreign protein within the target host cell, e.g., a bacterial, insect, yeast, plant, amphibian, reptile, avian, or mammalian cell, e.g., a CHO or HEK cell. Conveniently, manipulation of sequences and production of DNA for transformation may first performed in a bacterial host, e.g. E. coli, and usually vectors will include sequences to facilitate such manipulations, including a bacterial origin of replication and appropriate bacterial selection marker. Selection markers encode proteins necessary for the survival or growth of transformed host cells grown in a selective culture medium. Host cells not transformed with the vector containing the selection gene will not survive in the culture medium. Typical selection genes encode proteins that (a) confer resistance to antibiotics or other toxins, (b) complement auxotrophic deficiencies, or (c) supply critical nutrients not available from complex media. Exemplary vectors and methods for transformation of yeast are described, for example, in Burke, D., Dawson, D., & Stearns, T., Methods in yeast genetics:
a Cold Spring Harbor Laboratory course manual, Plainview, NY : Cold Spring Harbor Laboratory Press (2000). Expression vectors for use in the methods of the disclosure may include yeast or mammalian specific sequences, including a selectable auxotrophic or drug marker for identifying transformed host s. A drug marker may further be used to amplify copy number of the vector in a yeast host cell.
[0293] The polypeptide coding sequence of interest is operably linked to transcriptional and translational regulatory sequences that provide for expression of the polypeptide in the desired host cells, e.g., yeast or mammalian cells. These vector components may include, but are not limited to, one or more of the following: an enhancer element, a promoter, and a transcription termination sequence. Sequences for the secretion of the polypeptide may also be included, e.g. a signal sequence, and the like. An origin of replication, e.g., a yeast or mammalian origin of replication, is optional, as expression vectors may be integrated into the host cell genome.
[0294] Nucleic acids are "operably linked" when placed into a functional relationship with another nucleic acid sequence. For example, DNA for a signal sequence is operably linked to DNA for a polypeptide if it is expressed as a preprotein that participates in the secretion of the polypeptide; a promoter or enhancer is operably linked to a coding sequence if it affects the transcription of the sequence. Generally, "operably linked" means that the DNA sequences being linked are contiguous, and, in the case of a secretory leader, contiguous and in reading frame. However, enhancers do not have to be contiguous. Linking is accomplished by ligation at convenient restriction sites or via a PCR/recombination method familiar to those skilled in the art (GATEWAY® Technology (universal method for cloning DNA); Invitrogen, Carlsbad California). If such sites do not exist, the synthetic oligonucleotide adapters or linkers are used in accordance with conventional practice.
[0295] Promoters are untranslated sequences located upstream (5') to the start codon of a structural gene (generally within about 100 to 1000 bp) that control the transcription and translation of particular nucleic acid sequences to which they are operably linked. Such promoters fall into several classes: inducible, constitutive, and repressible promoters (that increase levels of transcription in response to absence of a repressor). Inducible promoters may initiate increased levels of transcription from DNA under their control in response to some change in culture conditions, e.g., the presence or absence of a nutrient or a change in temperature.
[0296] The promoter fragment may also serve as the site for homologous recombination and integration of the expression vector into the same site in the host cell, e.g., yeast or mammalian cell, genome; alternatively, a selectable marker may be used as the site for homologous recombination. Suitable promoters for use in different eukaryotic and prokaryotic cells are well known and commercially available.
[0297] The polypeptides of interest may be produced recombinantly not only directly, but also as a fusion polypeptide with a heterologous polypeptide, e.g. a signal sequence or other polypeptide
having a specific cleavage site at the N-terminus of the mature protein or polypeptide. In general, the signal sequence may be a component of the vector, or it may be a part of the polypeptide coding sequence that is inserted into the vector. The heterologous signal sequence selected preferably is one that is recognized and processed through one of the standard pathways available within the host cell, e.g., a mammalian cell, an insect cell, or a yeast cell. Additionally, these signal peptide sequences may be engineered to provide for enhanced secretion in expression systems. Secretion signals of interest also include mammalian and yeast signal sequences, which may be heterologous to the protein being secreted, or may be a native sequence for the protein being secreted. Signal sequences include pre peptide sequences, and in some instances may include propeptide sequences. Many such signal sequences are known in the art, including the signal sequences found on immunoglobulin chains, e.g., K28 preprotoxin sequence, PHA-E, FACE, human MCP-1, human serum albumin signal sequences, human Ig heavy chain, human Ig light chain, and the like. For example, see Hashimoto et. al., Protein Eng., 11(2):75 (1998); and Kobayashi et. al., Therapeutic Apheresis, 2(4):257 (1998)).
[0298] Transcription may be increased by inserting a transcriptional activator sequence into the vector. These activators are cis-acting elements of DNA, usually about from 10 to 300 bp, which act on a promoter to increase its transcription. Transcriptional enhancers are relatively orientation and position independent, having been found 5' and 3' to the transcription unit, within an intron, as well as within the coding sequence itself. The enhancer may be spliced into the expression vector at a position 5' or 3' to the coding sequence, but is preferably located at a site 5' from the promoter.
[0299] Expression vectors used in eukaryotic host cells may also contain sequences necessary for the termination of transcription and for stabilizing the rnRNA. Such sequences are commonly available from 3' to the translation termination codon, in untranslated regions of eukaryotic or viral DNAs or cDNAs. These regions contain nucleotide segments transcribed as polyadenylated fragments in the untranslated portion of the rnRNA.
[0300] Construction of suitable vectors containing one or more of the above -listed components employs standard ligation techniques or PCR/recombination methods. Isolated plasmids or DNA fragments are cleaved, tailored, and re-ligated in the form desired to generate the plasmids required or via recombination methods. For analysis to confirm correct sequences in plasmids constructed, the ligation mixtures are used to transform host cells, and successful transformants selected by antibiotic resistance (e.g. ampicillin or Zeocin) where appropriate. Plasmids from the transformants are prepared, analyzed by restriction endonuclease digestion, and/or sequenced.
[0301] As an alternative to restriction and ligation of fragments, recombination methods based on specific attachment (“ att ”) sites and recombination enzymes may be used to insert DNA sequences into a vector. Such methods are described, for example, by Landy, Ann. Rev. Biochem., 58:913-949 (1989); and are known to those of skill in the art. Such methods utilize intermolecular DNA recombination that is mediated by a mixture of lambda and E. coli -encoded recombination proteins.
Recombination occurs between att sites on the interacting DNA molecules. For a description of aft sites see Weisberg and Landy, Site-Specific Recombination in Phage Lambda, in Lambda II, P- 211- 250, Cold Spring Harbor, NY: Cold Spring Harbor Press (1983). The DNA segments flanking the recombination sites are switched, such that after recombination, the att sites are hybrid sequences comprised of sequences donated by each parental vector. The recombination can occur between DNAs of any topology.
[0302] Att sites may be introduced into a sequence of interest by ligating the sequence of interest into an appropriate vector; generating a PCR product containing att B sites through the use of specific primers; generating a cDNA library cloned into an appropriate vector containing att sites; and the like. [0303] Folding, as used herein, refers to the three-dimensional structure of polypeptides and proteins, where interactions between amino acid residues act to stabilize the structure. While non-covalent interactions are important in determining structure, usually the proteins of interest will have intra- and/or intermolecular covalent disulfide bonds formed by two cysteine residues. For naturally occurring proteins and polypeptides or derivatives and variants thereof, the proper folding is typically the arrangement that results in optimal biological activity, and can conveniently be monitored by assays for activity, e.g. ligand binding, enzymatic activity, etc.
[0304] In some instances, for example where the desired product is of synthetic origin, assays based on biological activity will be less meaningful. The proper folding of such molecules may be determined on the basis of physical properties, energetic considerations, modeling studies, etc.
[0305] The expression host may be further modified by the introduction of sequences encoding one or more enzymes that enhance folding and disulfide bond formation, i.e. foldases, chaperonins, etc. Such sequences may be constitutively or inducibly expressed in the host cell, using vectors, markers, etc. as known in the art. Preferably the sequences, including transcriptional regulatory elements sufficient for the desired pattern of expression, are stably integrated in the yeast genome through a targeted methodology.
[0306] For example, the eukaryotic protein disulfide isomerase (“PDI”) is not only an efficient catalyst of protein cysteine oxidation and disulfide bond isomerization, but also exhibits chaperone activity. Co-expression of PDI can facilitate the production of active proteins having multiple disulfide bonds. Also of interest is the expression of immunoglobulin heavy chain binding protein (“BIP”); cyclophilin; and the like.
[0307] Cultured mammalian cells are exemplary hosts for production of the disclosed anti-CoV-S antibodies and antigen-binding fragments thereof. As mentioned CHO cells are particularly suitable for expression of antibodies. Many procedures are known in the art for manufacturing monoclonal antibodies in mammalian cells. (See, Galfre, G. and Milstein, C., Methods Enzym., 73:3-46, 1981; Basalp et al, Turk. J. Biol., 24:189-196, 2000; Wurm, F.M., Nat. Biotechnol., 22:1393-1398, 2004; and Li et al, mAbs, 2(5):466-477, 2010). As mentioned in further detail infra, common host cell lines
employed in mammalian monoclonal antibody manufacturing schemes include, but are not limited to, human embryonic retinoblast cell line PER.C6® (Crucell N.V., Leiden, The Netherlands), NSO murine myeloma cells (Medical Research Council, London, UK), CV 1 monkey kidney cell line, 293 human embryonic kidney cell line, BHK baby hamster kidney cell line, VERO African green monkey kidney cell line, human cervical carcinoma cell line HELA, MDCK canine kidney cells, BRL buffalo rat liver cells, W138 human lung cells, HepG2 human liver cells, MMT mouse mammary tumor cells, TRI cells, MRC5 cells, Fs4 cells, myeloma or lymphoma cells, or Chinese Hamster ( Cricetulus griseus ) Ovary (CHO) cells, and the like. Many different subclones or sub-cell lines of CHO cells known in the art that are useful and optimized for production of recombinant monoclonal antibodies, such as the DP12 (CHO K1 dhfr-) cell line, NSO cells are a non-Ig secreting, non-light chain- synthesizing subclone of NS-1 cells that are resistant to azaguanine. Other Chinese Hamster and CHO cells are commercially available (from ATCC, etc.), including CHO-DXB11 (CHO-DUKX), CHO- pro3, CHO-DG44, CHO 1-15, CHO DP-12, Lec2, M1WT3, Lec8, μgsA-745, and the like, ah of which are genetically altered to optimize the cell line for various parameters. Monoclonal antibodies are commonly manufactured using a batch fed method whereby the monoclonal antibody chains are expressed in a mammalian cell line and secreted into the tissue culture medium in a bioreactor. Medium (or feed) is continuously supplied to the bioreactor to maximize recombinant protein expression. Recombinant monoclonal antibody is then purified from the collected media. In some circumstances, additional steps are needed to reassemble the antibodies through reduction of disulfide bonds, etc. Such production methods can be scaled to be as large as 10,000 L in a single batch or more. It is now routine to obtain as much as 20 μg/cell/day through the use of such cell lines and methodologies, providing titers as high as 10 g/L or more, amounting to 15 to 100 kg from bioreactors of 10 kL to 25 kL. (Li et ai, 2010). Various details of this production methodology, including cloning of the polynucleotides encoding the antibodies into expression vectors, transfecting cells with these expression vectors, selecting for transfected cells, and expressing and purifying the recombinant monoclonal antibodies from these cells are provided below.
[0308] For recombinant production of an anti-CoV-S antibody or antigen-binding fragment in mammalian cells, nucleic acids encoding the antibody or fragment thereof are generally inserted into a replicable vector for further cloning (amplification of the DNA) or for expression. DNA encoding the antibody is readily isolated or synthesized using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to DNAs encoding the heavy and light chains of the antibody). The vector components generally include, but are not limited to, one or more of the following: a signal sequence, an origin of replication, one or more marker genes, an enhancer element, a promoter, and a transcription termination sequence. Selection of promoters, terminators, selectable markers, vectors, and other elements is a matter of routine design within the level of
ordinary skill in the art. Many such elements are known in the art and are available through commercial suppliers.
[0309] The antibodies of this disclosure may be produced recombinantly not only directly, but also as a fusion polypeptide with a heterologous polypeptide, which is preferably a signal sequence or other polypeptide having a specific cleavage site at the N-terminus of the mature protein or polypeptide.
The homologous or heterologous signal sequence selected preferably is one that is recognized and processed (i.e., cleaved by a signal peptidase) by the host cell. In mammalian cell expression, mammalian signal sequences as well as viral secretory leaders, for example, the herpes simplex gD signal, are available.
[0310] Such expression vectors and cloning vectors will generally contain a nucleic acid sequence that enables the vector to replicate in one or more selected host cells. Typically, in cloning vectors this sequence is one that enables the vector to replicate independently of the host chromosomal DNA, and includes origins of replication or autonomously replicating sequences. Such sequences are well known for a variety of bacteria, yeast, and viruses, e.g. , the origin of replication from the plasmid pBR322 is suitable for most Gram-negative bacteria, the 2mu plasmid origin is suitable for yeast, and various viral origins (Simian Virus 40 (“SV40”), polyoma, adenovirus, vesicular stomatitis virus (“VSV”), or bovine papillomavirus (“BPV”) are useful for cloning vectors in mammalian cells. Generally, the origin of replication component is not needed for mammalian expression vectors (the SV40 origin may typically be used only because it contains the early promoter).
[0311] These vectors will also typically contain a selection gene, also termed a selectable marker. Typical selection genes encode proteins that (a) confer resistance to antibiotics or other toxins, e.g., ampicillin, neomycin, methotrexate, or tetracycline, (b) complement auxotrophic deficiencies, or (c) supply critical nutrients not available from complex media, e.g., the gene encoding D-alanine racemase for Bacilli.
[0312] One example of a selection scheme utilizes a drug to arrest growth of a host cell. Drug selection is generally used to select for cultured mammalian cells into which foreign DNA has been inserted. Such cells are commonly referred to as "transfectants". Cells that have been cultured in the presence of the selective agent and are able to pass the gene of interest to their progeny are referred to as "stable transfectants." Examples of such dominant selection use the drugs neomycin, mycophenolic acid, and hygromycin. An exemplary selectable marker is a gene encoding resistance to the antibiotic neomycin. Selection is carried out in the presence of a neomycin-type drug, such as G-418 or the like. Those cells that are successfully transformed with a heterologous gene produce a protein conferring drug resistance and thus survive the selection regimen.
[0313] Selection systems can also be used to increase the expression level of the gene of interest, a process referred to as "amplification." Amplification of transfectants typically occurs by culturing the cells in the presence of a low level of the selective agent and then increasing the amount of selective
agent to select for cells that produce high levels of the products of the introduced genes. Exemplary suitable selectable markers for mammalian cells are those that enable the identification of cells competent to take up the antibody nucleic acid, such as dihydrofolate reductase (“DHFR”), thymidine kinase, metallothionein-I and -II, preferably primate metallothionein genes, adenosine deaminase, ornithine decarboxylase, etc.
[0314] For example, an amplifiable selectable marker for mammalian cells is dihydrofolate reductase, which confers resistance to methotrexate. Other drug resistance genes (e.g. hygromycin resistance, multi-drug resistance, puromycin acetyltransferase) can also be used. Cells transformed with the DHFR selection gene are first identified by culturing all of the transformants in a culture medium that contains methotrexate (“MTX”), a competitive antagonist of DHFR. An appropriate host cell when wild-type DHFR is employed is the Chinese hamster ovary (“CHO”) cell line deficient in DHFR activity.
[0315] Alternatively, host cells (particularly wild-type hosts that contain endogenous DHFR) transformed or co-transformed with DNA sequences encoding antibody, wild-type DHFR protein, and another selectable marker such as aminoglycoside 3'-phosphotransferase (“APH”) can be selected by cell growth in medium containing a selection agent for the selectable marker such as an aminoglycosidic antibiotic, e.g., kanamycin, neomycin, or G-418. See U.S. Patent No. 4,965,199. [0316] These vectors may comprise an enhancer sequence that facilitates transcription of a DNA encoding the antibody. Many enhancer sequences are known from mammalian genes (for example, globin, elastase, albumin, alpha-fetoprotein, and insulin). A frequently used enhancer is one derived from a eukaryotic cell virus. Examples thereof include the SV40 enhancer on the late side of the replication origin (bp 100-270), the cytomegalovirus early promoter enhancer, the polyoma enhancer on the late side of the replication origin, and adenovirus enhancers (See also Yaniv, Nature, 297:17- 18 (1982) on enhancing elements for activation of eukaryotic promoters). The enhancer may be spliced into the vector at a position 5' or 3' to the antibody-encoding sequence, but is preferably located at a site 5' from the promoter.
[0317] Expression and cloning vectors will also generally comprise a promoter that is recognized by the host organism and is operably linked to the antibody nucleic acid. Promoter sequences are known for eukaryotes. Virtually all eukaryotic genes have an AT-rich region located approximately 25 to 30 bases upstream from the site where transcription is initiated. Another sequence found 70 to 80 bases upstream from the start of transcription of many genes is a CNCAAT region where N may be any nucleotide. At the 3' end of most eukaryotic genes is an AATAAA sequence that may be the signal for addition of the poly A tail to the 3' end of the coding sequence. All of these sequences are suitably inserted into eukaryotic expression vectors.
[0318] Antibody transcription from vectors in mammalian host cells may be controlled, for example, by promoters obtained from the genomes of viruses such as polyoma virus, fowlpox virus, adenovirus
(such as Adenovirus 2), BPV, avian sarcoma virus, cytomegalovirus, a retrovirus, hepatitis-B virus, and most preferably SV40, from heterologous mammalian promoters, e.g., the actin promoter or an immunoglobulin promoter, from heat-shock promoters, provided such promoters are compatible with the host cell systems.
[0319] The early and late promoters of the SV40 virus are conveniently obtained as an SV40 restriction fragment that also contains the SV40 viral origin of replication. The immediate early promoter of the human cytomegalovirus is conveniently obtained as a Hindlll E restriction fragment. A system for expressing DNA in mammalian hosts using the BPV as a vector is disclosed in U.S. Patent No. 4,419,446. A modification of this system is described in U.S. Patent No. 4,601,978. See also Reyes et ai, Nature, 297:598-601 (1982) on expression of human beta-interferon cDNA in mouse cells under the control of a thymidine kinase promoter from herpes simplex virus. Alternatively, the Rous sarcoma virus long terminal repeat can be used as the promoter.
[0320] Strong transcription promoters can be used, such as promoters from SV40, cytomegalovirus, or myeloproliferative sarcoma virus. See, e.g., U.S. Patent No. 4,956,288 and U.S. Patent Publication No. 20030103986. Other suitable promoters include those from metallothionein genes (U.S. Patent Nos. 4,579,821 and 4,601,978) and the adenovirus major late promoter. Expression vectors for use in mammalian cells include pZP-1, pZP-9, and pZMP21, which have been deposited with the American Type Culture Collection, 10801 University Blvd., Manassas, VA. USA under accession numbers 98669, 98668, and PTA-5266, respectively, and derivatives of these vectors.
[0321] Expression vectors used in eukaryotic host cells (yeast, fungus, insect, plant, animal, human, or a nucleated cell from other multicellular organism) will also generally contain sequences necessary for the termination of transcription and for stabilizing the rnRNA. Such sequences are commonly available from the 5' and, occasionally 3', untranslated regions of eukaryotic or viral DNAs or cDNAs. These regions contain nucleotide segments transcribed as polyadenylated fragments in the untranslated portion of the rnRNA encoding the antibody. One useful transcription termination component is the bovine growth hormone polyadenylation region. See WO 94/11026 and the expression vector disclosed therein.
[0322] Suitable host cells for cloning or expressing the subject antibodies include prokaryote, yeast, or higher eukaryote cells described above. However, interest has been greatest in vertebrate cells, and propagation of vertebrate cells in culture has become a routine procedure. Examples of useful mammalian host cell lines are monkey kidney CV1 line transformed by SV40 (COS-1 (ATCC No. CRL 1650); and COS-7, ATCC CRL 1651); human embryonic kidney line (293 or 293 cells subcloned for growth in suspension culture, (ATCC No. CRL 1573; Graham et ai, J. Gen. Virol., 36:59-72 (1977)); baby hamster kidney cells (BHK, ATCC CCL 10, ATCC No. CRL 1632; BHK 570, ATCC No. CRL 10314); CHO cells (CHO-K1, ATCC No. CCL 61; CHO-DG44, Urlaub et ai, Proc. Natl. Acad. Sci. USA, 77:4216-4220 (1980)); mouse sertoli cells (TM4, Mather, Biol. Reprod.,
23:243-251 (1980)); monkey kidney cells (CV1 ATCC CCL 70); African green monkey kidney cells (VERO-76, ATCC CRL-1587); human cervical carcinoma cells (HELA, ATCC CCL 2); canine kidney cells (MDCK, ATCC CCL 34); buffalo rat liver cells (BRL 3A, ATCC CRL 1442); human lung cells (W138, ATCC CCL 75); human liver cells (Hep G2, HB 8065); mouse mammary tumor (MMT 060562, ATCC CCL51); TRI cells (Mather et al, Annals N.Y. Acad. Sci., 383:44-68 (1982)); MRC 5 cells; LS4 cells; and a human hepatoma line (Hep G2). Additional suitable cell lines are known in the art and available from public depositories such as the American Type Culture Collection, Manassas, VA.
[0323] Host cells are transformed with the above -described expression or cloning vectors for antibody production and cultured in conventional nutrient media modified as appropriate for inducing promoters, selecting transformants, or amplifying the genes encoding the desired sequences as discussed supra.
[0324] The mammalian host cells used to produce the antibody of this disclosure may be cultured in a variety of media. Commercially available media such as Ham's L10 (Sigma-Aldrich Corporation, St. Louis, MO), Minimal Essential Medium ((“MEM” (Sigma-Aldrich Corporation, St. Louis, MO), Roswell Park Memorial Institute- 1640 medium (“RPMI-1640”, Sigma-Aldrich Corporation, St.
Louis, MO), and Dulbecco's Modified Eagle's Medium ((“DMEM” Sigma-Aldrich Corporation, St. Louis, MO) are suitable for culturing the host cells. In addition, any of the media described in Ham et al, Meth. Enz·, 58:44 (1979), Barnes et aI., AhaI. Biochem., 102:255 (1980), U.S. Patent Nos. 4,767,704; 4,657,866; 4,927,762; 4,560,655; or 5,122,469; WO 90/03430; WO 87/00195; or U.S. Patent Reexam No. 30,985 can be used as culture media for the host cells. Any of these media may be supplemented as necessary with hormones and/or other growth factors (such as insulin, transferrin, or epidermal growth factor), salts (such as sodium chloride, calcium, magnesium, and phosphate), buffers (such as HEPES), nucleotides (such as adenosine and thymidine), antibiotics (such as Gentamycin drug), trace elements (defined as inorganic compounds usually present at final concentrations in the micromolar range), and glucose or an equivalent energy source. Any other necessary supplements may also be included at appropriate concentrations that would be known to those skilled in the art. The culture conditions, such as temperature, pH, and the like, are those previously used with the host cell selected for expression, and will be apparent to the ordinarily skilled artisan. Methods of development and optimization of media and culture conditions are known in the art (See, Gronemeyer et al., Bioengineering, 1(4): 188-212, 2014).
[0325] After culture conditions are optimized and a preferred cell line clone is selected, these cells are cultured (either adherent cells or suspension cultures) most typically in a batch-fed process in a bioreactor (many models are commercially available) that involves continuously feeding the cell culture with medium and feed, optimized for the particular cell line chosen and selected for this purpose. (See, Butler, M., Appl. Microbiol. BiotechnoL, 68:283-291, 2005; and Kelley, B., mAb,
l(5):443-452, 2009). Perfusion systems are also available in which media and feed are continuously supplied to the culture while the same volume of media is being withdrawn from the bioreactor. (Wurm, 2004). Synthetic media, also commercially available, are available for growing cells in a batch-fed culture, avoiding the possibility of contamination from outside sources, such as with the use of animal components, such as bovine serum albumin, etc. However, animal-component-free hydrolysates are commercially available to help boost cell density, culture viability and productivity. (Li et al, 2010). Many studies have been performed in an effort to optimize cell culture media, including careful attention to head space available in roller bottles, redox potentials during growth and expression phases, presence of reducing agents to maintain disulfide bonds during production, etc. (See, for instance, Hutterer et al, mAbs, 5(4):608-613, 2013; and Mullan et ai, BMC Proceed., 5(Suppl 8):P110, 2011). Various methodologies have been developed to address the possibility of harmful oxidation during recombinant monoclonal antibody production. (See, for example, U.S.
Patent No. 8,574,869). Cultured cells may be grown by feeding nutrients continuously or as separately administered amounts. Often various process parameters such as cell concentration, pH, temperature, CO2, d02, osmolality, amount of metabolites such as glucose, lactate, glutamine and glutamate, and the like, are monitored by the use of probes during the cell growth either on-line by direct connection to calibrated analyzers or off-line by intervention of operators. The culturing step also typically involves ensuring that the cells growing in culture maintain the transfected recombinant genes by any means known in the art for cell selection.
[0326] Following fermentation, i.e. , upon reaching maximum cell growth and recombinant protein expression, the culturing step is typically followed by a harvesting step, whereby the cells are separated from the medium and a harvested cell culture media is thereby obtained. (See, Liu et al. , mAbs, 2(5):480-499, 2010). Typically, various purification steps, involving column chromatography and the like, follow culturing to separate the recombinant monoclonal antibody from cell components and cell culture media components. The exact purification steps needed for this phase of the production of recombinant monoclonal antibodies depends on the site of expression of the proteins, i.e., in the cytosol of the cells themselves, or the more commonly preferred route of protein excreted into the cell culture medium. Various cell components may be separated using techniques known in the art such as differential centrifugation techniques, gravity-based cell settling, and/or size exclusion chromatograph/filtration techniques that can include tangential flow micro-filtration or depth filtration. (See, Pollock et al. , Biotechnol. Bioeng. , 110:206-219, 2013, and Liu et al. , 2010). Centrifugation of cell components may be achieved on a large scale by use of continuous disk stack centrifuges followed by clarification using depth and membrane filters. (See, Kelley, 2009). Most often, after clarification, the recombinant protein is further purified by Protein A chromatography due to the high affinity of Protein A for the Fc domain of antibodies, and typically occurs using a low pH/acidification elution step (typically the acidification step is combined with a precautionary virus
inactivation step). Flocculation and/or precipitation steps using acidic or cationic polyelectrolytes may also be employed to separate animal cells in suspension cultures from soluble proteins. (Liu et al, 2010). Lastly, anion- and cation-exchange chromatography, hydrophobic interaction chromatograph (“HIC”), hydrophobic charge induction chromatograph (HCIC), hydroxyapatite chromatography using ceramic hydroxyapatite (Cas(P04)30H)2, and combinations of these techniques are typically used to polish the solution of recombinant monoclonal antibody. Final formulation and concentration of the desired monoclonal antibody may be achieved by use of ultracentrifugation techniques. Purification yields are typically 70 to 80%. (Kelley, 2009).
[0327] The terms "desired protein" or "desired antibody" herein are used interchangeably and refer generally to a parent antibody specific to a target, i.e., CoV-S or a chimeric or humanized antibody or a binding portion thereof derived therefrom as described herein. The term “antibody” is intended to include any polypeptide chain-containing molecular structure with a specific shape that fits to and recognizes an epitope, where one or more non-covalent binding interactions stabilize the complex between the molecular structure and the epitope. The archetypal antibody molecule is the immunoglobulin, and ah types of immunoglobulins, IgG, IgM, IgA, IgE, IgD, etc., from ah sources, e.g. human, rodent, rabbit, cow, sheep, pig, dog, other mammals, chicken, other avians, etc., are considered to be “antibodies.” Examples thereof include chimeric antibodies, human antibodies and other non-human mammalian antibodies, humanized antibodies, single chain antibodies (such as scFvs), camelbodies, nanobodies, IgNAR (single-chain antibodies which may be derived from sharks, for example), small-modular immunopharmaceuticals (“SMIPs”), and antibody fragments such as Fabs, Fab', F(ab')2, and the like ( See Streltsov et ai, Protein ScL, 14(11) :2901 -9 (2005); Greenberg et al, Nature, 374(6518):168-73 (1995); Nuttall et al, Mol. Immunol., 38(4):313-26 (2001); Flamers- Casterman et al., Nature, 363(6428):446-8 (1993); Gill et al., Curr. Opin. Biotechnol., (6):653-8 (2006)).
[0328] For example, antibodies or antigen-binding fragments thereof may be produced by genetic engineering. In this technique, as with other methods, antibody-producing cells are sensitized to the desired antigen or immunogen. The messenger RNA isolated from antibody producing cells is used as a template to make cDNA using PCR amplification. A library of vectors, each containing one heavy chain gene and one light chain gene retaining the initial antigen specificity, is produced by insertion of appropriate sections of the amplified immunoglobulin cDNA into the expression vectors. A combinatorial library is constructed by combining the heavy chain gene library with the light chain gene library. This results in a library of clones that co-express a heavy and light chain (resembling the Fab fragment or antigen-binding fragment of an antibody molecule). The vectors that carry these genes are co-transfected into a host cell. When antibody gene synthesis is induced in the transfected host, the heavy and light chain proteins self-assemble to produce active antibodies that can be detected by screening with the antigen or immunogen.
[0329] Antibody coding sequences of interest include those encoded by native sequences, as well as nucleic acids that, by virtue of the degeneracy of the genetic code, are not identical in sequence to the disclosed nucleic acids, and variants thereof. Variant polypeptides can include amino acid (“aa”) substitutions, additions, or deletions. The amino acid substitutions can be conservative amino acid substitutions or substitutions to eliminate non-essential amino acids, such as to alter a glycosylation site, or to minimize misfolding by substitution or deletion of one or more cysteine residues that are not necessary for function. Variants can be designed so as to retain or have enhanced biological activity of a particular region of the protein (e.g., a functional domain, catalytic amino acid residues, etc). Variants also include fragments of the polypeptides disclosed herein, particularly biologically active fragments and/or fragments corresponding to functional domains. Techniques for in vitro mutagenesis of cloned genes are known. Also included in the subject disclosure are polypeptides that have been modified using ordinary molecular biological techniques so as to improve their resistance to proteolytic degradation or to optimize solubility properties or to render them more suitable as a therapeutic agent.
[0330] Chimeric antibodies may be made by recombinant means by combining the VL and VH regions, obtained from antibody producing cells of one species with the constant light and heavy chain regions from another. Typically, chimeric antibodies utilize rodent or rabbit variable regions and human constant regions, in order to produce an antibody with predominantly human domains.
The production of such chimeric antibodies is well known in the art, and may be achieved by standard means (as described, e.g., in U.S. Patent No. 5,624,659, incorporated herein by reference in its entirety). It is further contemplated that the human constant regions of chimeric antibodies of the disclosure may be selected from IgGl, IgG2, IgG3, and IgG4 constant regions.
[0331] Humanized antibodies are engineered to contain even more human-like mmunoglobulin domains, and incorporate only the complementarity determining regions of the animal -derived antibody. This is accomplished by carefully examining the sequence of the hyper- variable loops of the variable regions of the monoclonal antibody and fitting them to the structure of the human antibody chains. Although facially complex, the process is straightforward in practice. See, e.g., U.S. Patent No. 6,187,287, incorporated fully herein by reference.
[0332] In addition to entire immunoglobulins (or their recombinant counterparts), immunoglobulin fragments comprising the epitope binding site (e.g., Fab’, F(ab’)2, or other fragments) may be synthesized. “Fragment” or minimal immunoglobulins may be designed utilizing recombinant immunoglobulin techniques. For instance, “Fv” immunoglobulins for use in the present disclosure may be produced by synthesizing a fused variable light chain region and a variable heavy chain region. Combinations of antibodies are also of interest, e.g. diabodies, which comprise two distinct Fv specificities. In another embodiment, small molecule immunopharmaceuticals (“SMIPs”), camelbodies, nanobodies, and IgNAR are encompassed by immunoglobulin fragments.
[0333] Immunoglobulins and fragments thereof may be modified post-translationally, e.g. to add effector moieties such as chemical linkers, detectable moieties, such as fluorescent dyes, enzymes, toxins, substrates, bioluminescent materials, radioactive materials, chemiluminescent moieties, and the like, or specific binding moieties, such as streptavidin, avidin, or biotin, and the like may be utilized in the methods and compositions of the present disclosure. Examples of additional effector molecules are provided infra.
[0334] A polynucleotide sequence "corresponds" to a polypeptide sequence if translation of the polynucleotide sequence in accordance with the genetic code yields the polypeptide sequence (i.e., the polynucleotide sequence "encodes" the polypeptide sequence), one polynucleotide sequence "corresponds" to another polynucleotide sequence if the two sequences encode the same polypeptide sequence.
[0335] A "heterologous" region or domain of a DNA construct is an identifiable segment of DNA within a larger DNA molecule that is not found in association with the larger molecule in nature.
Thus, when the heterologous region encodes a mammalian gene, the DNA flanking the gene usually does not flank the mammalian genomic DNA in the genome of the source organism. Another example of a heterologous region is a construct where the coding sequence itself is not found in nature (e.g., a cDNA where the genomic coding sequence contains introns or synthetic sequences having codons different than the native gene). Allelic variations or naturally-occurring mutational events do not give rise to a heterologous region of DNA as defined herein.
[0336] A "coding sequence" is an in-frame sequence of codons that correspond to or encode a protein or peptide sequence. Two coding sequences correspond to each other if the sequences or their complementary sequences encode the same amino acid sequences. A coding sequence in association with appropriate regulatory sequences may be transcribed and translated into a polypeptide. A polyadenylation signal and transcription termination sequence will usually be located 3' to the coding sequence. A "promoter sequence" is a DNA regulatory region capable of initiating transcription of a downstream (3' direction) coding sequence, and typically contain additional sites for binding of regulatory molecules, e.g., transcription factors, that affect the transcription of the coding sequence. A coding sequence is "under the control" of the promoter sequence or "operatively linked" to the promoter when RNA polymerase binds the promoter sequence in a cell and transcribes the coding sequence into mRNA, which is then in turn translated into the protein encoded by the coding sequence.
[0337] The general structure of antibodies in vertebrates now is well understood. See Edelman, G. M., Ann. N.Y. Acad. Sci., 190:5 (1971). Antibodies consist of two identical light polypeptide chains of molecular weight approximately 23,000 daltons (the “light chain”), and two identical heavy chains of molecular weight 53,000-70,000 (the “heavy chain”). The four chains are joined by disulfide bonds in a “Y” configuration wherein the light chains bracket the heavy chains starting at the mouth of the “Y”
configuration. The “branch” portion of the “Y” configuration is designated the Fab region; the stem portion of the “Y” configuration is designated the Fc region. The amino acid sequence orientation runs from the N-terminal end at the top of the “Y” configuration to the C-terminal end at the bottom of each chain. The N-terminal end possesses the variable region having specificity for the antigen that elicited it, and is approximately 100 amino acids in length, there being slight variations between light and heavy chain and from antibody to antibody.
[0338] The variable region is linked in each chain to a constant region that extends the remaining length of the chain and that within a particular class of antibody does not vary with the specificity of the antibody (i.e., the antigen eliciting it). There are five known major classes of constant regions that determine the class of the immunoglobulin molecule (IgG, IgM, IgA, IgD, and IgE corresponding to g, m, a, d, and e (gamma, mu, alpha, delta, or epsilon) heavy chain constant regions). The constant region or class determines subsequent effector function of the antibody, including activation of complement ( see Kabat, E. A., Structural Concepts in Immunology and Immunochemistry, 2nd Ed., p. 413-436, New York, NY: Flolt, Rinehart, Winston (1976)), and other cellular responses ( see Andrews et ai, Clinical Immunology, pp. 1-18, W. B. Sanders, Philadelphia, PA (1980); Kohl et ai, Immunology, 48:187 (1983)); while the variable region determines the antigen with which it will react. Light chains are classified as either k (kappa) or l (lambda). Each heavy chain class can be prepared with either kappa or lambda light chain. The light and heavy chains are covalently bonded to each other, and the “tail” portions of the two heavy chains are bonded to each other by covalent disulfide linkages when the immunoglobulins are generated either by hybridomas or by B -cells.
[0339] The expression “variable region” or “VR” refers to the domains within each pair of light and heavy chains in an antibody that are involved directly in binding the antibody to the antigen. Each heavy chain has at one end a variable region (VFI) followed by a number of constant domains. Each light chain has a variable region (VL) at one end and a constant domain at its other end; the constant domain of the light chain is aligned with the first constant domain of the heavy chain, and the light chain variable domain is aligned with the variable domain of the heavy chain.
[0340] The expressions “complementarity-determining region,” “hypervariable region,” or “CDR” refer to one or more of the hyper-variable or complementarity-determining regions (“CDRs”) found in the variable regions of light or heavy chains of an antibody ( See Kabat et ai, Sequences of Proteins of Immunological Interest, 4th ed., Bethesda, MD: U.S. Dept of Flealth and Human Services, Public Health Service, National Institutes of Health (1987)). These expressions include the hypervariable regions as defined by Kabat et ai, ( Sequences of Proteins of Immunological Interest, NIH Publication No. 91-3242, Bethesda, MD: U.S. Dept of Health and Human Services, National Institutes of Health (1983)) or the hypervariable loops in 3-dimensional structures of antibodies (Chothia and Lesk, J.
Mol. Biol., 196:901-917 (1987)). The CDRs in each chain are held in close proximity by framework regions (“FRs”) and, with the CDRs from the other chain, contribute to the formation of the antigen
binding site. Within the CDRs there are select amino acids that have been described as the selectivity determining regions (“SDRs”) that represent the critical contact residues used by the CDR in the antibody-antigen interaction ( see Kashmiri et al, Methods, 36(l):25-34 (2005)).
[0341] An "epitope" or "binding site" is an area or region on an antigen to which an antigen-binding peptide (such as an antibody) specifically binds. A protein epitope may comprise amino acid residues directly involved in the binding (also called immunodominant component of the epitope) and other amino acid residues, which are not directly involved in the binding, such as amino acid residues that are effectively blocked by the specifically antigen binding peptide (in other words, the amino acid residue is within the "footprint" of the specifically antigen binding peptide). The term epitope herein includes both types of amino acid binding sites in any particular region of CoV-S, e.g., SARS-CoV-S or SARS-CoV-2-S, that specifically binds to an anti-CoV-S antibody. CoV-S may comprise a number of different epitopes, which may include, without limitation, (1) linear peptide antigenic determinants, (2) conformational antigenic determinants that consist of one or more non-contiguous amino acids located near each other in a mature CoV-S conformation; and (3) post-translational antigenic determinants that consist, either in whole or part, of molecular structures covalently attached to a CoV-S protein such as carbohydrate groups. In particular, the term “epitope” includes the specific residues in a protein or peptide, e.g., CoV-S, which are involved in the binding of an antibody to such protein or peptide as determined by known and accepted methods such as alanine scanning techniques or the use of various S protein portions with varying lengths.
[0342] The phrase that an antibody (e.g., first antibody) binds "substantially" or "at least partially" the same epitope as another antibody (e.g., second antibody) means that the epitope binding site for the first antibody comprises at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more of the amino acid residues on the antigen that constitutes the epitope binding site of the second antibody. Also, that a first antibody binds substantially or partially the same or overlapping epitope as a second antibody means that the first and second antibodies compete in binding to the antigen, as described above. Thus, the term "binds to substantially the same epitope or determinant as" a monoclonal antibody means that an antibody "competes" with the antibody.
[0343] The phrase "binds to the same or overlapping epitope or determinant as" an antibody of interest means that an antibody "competes" with said antibody of interest for at least one, (e.g., at least 2, at least 3, at least 4, at least 5) or all residues on CoV-S to which said antibody of interest specifically binds. The identification of one or more antibodies that bind(s) to substantially or essentially the same epitope as the monoclonal antibodies described herein can be readily determined using alanine scanning. Additionally, any one of variety of immunological screening assays in which antibody competition can be assessed. A number of such assays are routinely practiced and well known in the art (see, e.g., U.S. Patent No. 5,660,827, issued Aug. 26, 1997, which is specifically incorporated herein by reference). It will be understood that actually determining the epitope to which
an antibody described herein binds is not in any way required to identify an antibody that binds to the same or substantially the same or overlapping epitope as the monoclonal antibody described herein. [0344] For example, where the test antibodies to be examined are obtained from different source animals, or are even of a different Ig isotype, a simple competition assay may be employed in which the control antibody is mixed with the test antibody and then applied to a sample containing CoV-S. Protocols based upon ELISAs, radioimmunoassays, Western blotting, and the use of BIACORE®
(GE Healthcare Life Sciences, Marlborough, MA) analysis are suitable for use in such simple competition studies.
[0345] In certain embodiments, the control anti-CoV-S antibody is pre-mixed with varying amounts of the test antibody (e.g., in ratios of about 1:1, 1:2, 1:10, or about 1:100) for a period of time prior to applying to the CoV-S (e.g., SARS-CoV-S or SARS-CoV-2-S) antigen sample. In other embodiments, the control and varying amounts of test antibody can simply be added separately and admixed during exposure to the SARS-CoV-S or SARS-CoV-2-S antigen sample. As long as bound antibodies can be distinguished from free antibodies (e.g., by using separation or washing techniques to eliminate unbound antibodies) and control antibody from the test antibody (e.g., by using species specific or isotype specific secondary antibodies or by specifically labeling the control antibody with a detectable label) it can be determined if the test antibody reduces the binding of the control antibody to the SARS-CoV-S or SARS-CoV-2-S antigens, indicating that the test antibody recognizes substantially the same epitope as the control anti-CoV-S antibody. The binding of the (labeled) control antibody in the presence of a completely irrelevant antibody (that does not bind CoV-S) can serve as the control high value. The control low value can be obtained by incubating the labeled control antibody with the same but unlabeled control antibody, where competition would occur and reduce binding of the labeled antibody. In a test assay, a significant reduction in labeled antibody reactivity in the presence of a test antibody is indicative of a test antibody that recognizes substantially the same epitope, i.e., one that competes with the labeled control antibody. For example, any test antibody that reduces the binding of the control antibody to SARS-CoV-S or SARS-CoV-2-S by at least about 50%, such as at least about 60%, or more preferably at least about 70% (e.g., about 65-100%), at any ratio of test antibody between about 1:1 or 1:10 and about 1:100 is considered to be an antibody that binds to substantially the same or overlapping epitope or determinant as the control antibody.
[0346] Preferably, such test antibody will reduce the binding of the control antibody to SARS-CoV-S or SARS-CoV-2-S (or another CoV-S) antigen preferably at least about 50%, at least about 60%, at least about 80%, or at least about 90% (e.g., about 95%) of the binding of the control antibody observed in the absence of the test antibody.
[0347] A simple competition assay in which a test antibody is applied at saturating concentration to a surface onto which SARS-CoV-S or SARS-CoV-2-S (or another CoV-S) is immobilized also may be
advantageously employed. The surface in the simple competition assay is preferably a BIACORE® (GE Healthcare Life Sciences, Marlborough, MA) chip (or other media suitable for surface plasmon resonance (“SPR”) analysis). The binding of a control antibody that binds SARS-CoV-S or SARS- CoV-2-S to the COV-S-coated surface is measured. This binding to the SARS-CoV-S- or SARS- CoV -2 -S -containing surface of the control antibody alone is compared with the binding of the control antibody in the presence of a test antibody. A significant reduction in binding to the SARS-CoV-S- or S ARS -Co V -2 -S -containing surface by the control antibody in the presence of a test antibody indicates that the test antibody recognizes substantially the same epitope as the control antibody such that the test antibody "competes" with the control antibody. Any test antibody that reduces the binding of control antibody by at least about 20% or more, at least about 40%, at least about 50%, at least about 70%, or more, can be considered to be an antibody that binds to substantially the same epitope or determinant as the control antibody. Preferably, such test antibody will reduce the binding of the control antibody to SARS-CoV-S or SARS-CoV-2-S by at least about 50% (e.g., at least about 60%, at least about 70%, or more). It will be appreciated that the order of control and test antibodies can be reversed; i.e. the control antibody can be first bound to the surface and then the test antibody is brought into contact with the surface thereafter in a competition assay. Preferably, the “sandwich- style” binding assay infra is used. Alternatively, the antibody having greater affinity for SARS-CoV-S or SARS-CoV-2-S antigen is bound to the SARS-CoV-S- or SARS-CoV-2-S-containing surface first, as it will be expected that the decrease in binding seen for the second antibody (assuming the antibodies are competing) will be of greater magnitude. Further examples of such assays are provided in e.g., Saunal and Regenmortel, J. Immunol. Methods, 183:33-41 (1995), the disclosure of which is incorporated herein by reference.
[0348] In addition, whether an antibody binds the same or overlapping epitope(s) on COV-S as another antibody or the epitope bound by a test antibody may in particular be determined using a Western-blot based assay. In this assay a library of peptides corresponding to the antigen bound by the antibody, the CoV-S protein, is made, that comprise overlapping portions of the protein, typically 10- 25, 10-20, or 10-15 amino acids long. These different overlapping amino acid peptides encompassing the CoV-S sequence are synthesized and covalently bound to a PEPSPOTS™ nitrocellulose membrane (JPT Peptide Technologies, Berlin, Germany). Blots are then prepared and probed according to the manufacturer's recommendations.
[0349] Essentially, the immunoblot assay then detects by fluorometric means what peptides in the library bind to the test antibody and thereby can identify what residues on the antigen, i.e., COV-S, interact with the test antibody. (See U.S. Patent No. 7,935,340, incorporated by reference herein). [0350] Various epitope mapping techniques are known in the art. By way of example, X-ray co- crystallography of the antigen and antibody; NMR; SPR (e.g., at 25° or 37°C); array-based oligo peptide scanning (or “pepscan analysis”); site -directed mutagenesis (e.g., alanine scanning);
mutagenesis mapping; hydrogen-deuterium exchange; phage display; and limited proteolysis are all epitope mapping techniques that are well known in the art (See, e.g., Epitope Mapping Protocols: Second Edition, Methods in Molecular Biology, , editors Mike Schutkowski and Ulrich Reineke, 2nd Ed., New York, NY: Humana Press (2009), and Epitope Mapping Protocols, Methods in Molecular Biology, editor Glenn Morris, 1st Ed., New York, NY: Humana Press (1996), both of which are herein incorporated by referenced in their entirety).
[0351] The identification of one or more antibodies that bind(s) to substantially or essentially the same epitope as the monoclonal antibodies described herein, e.g., any one of antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, ADI-58131, can be readily determined using any one of variety of immunological screening assays in which antibody competition can be assessed. A number of such assays are routinely practiced and well known in the art (see, e.g., U.S. Patent No. 5,660,827, issued Aug. 26, 1997, which is incorporated herein by reference). It will be understood that determining the epitope to which an antibody described herein binds is not in any way required to identify an antibody that binds to the same or substantially the same epitope as the monoclonal antibody described herein.
[0352] For example, where the test antibodies to be examined are obtained from different source animals, or are even of a different Ig isotype, a simple competition assay may be employed in which the control antibody (one of antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, ADI-58131, for example) is mixed with the test antibody and then applied to a sample containing either or both SARS-CoV-S or SARS-CoV-2-S, each of which is known to be bound by antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, ADI-58131. Protocols based upon ELISAs, radioimmunoassays, Western blotting, and BIACORE® (GE Healthcare Life Sciences, Marlborough, MA) analysis (as described in the Examples section herein) are suitable for use in such simple competition studies.
[0353] In certain embodiments, the method comprises pre-mixing the control antibody with varying amounts of the test antibody (e.g., in ratios of about 1:1, 1:2, 1:10, or about 1:100) for a period of time prior to applying to the CoV-S antigen sample. In other embodiments, the control and varying amounts of test antibody can be added separately and admixed during exposure to the CoV-S antigen sample. As long as bound antibodies can be distinguished from free antibodies (e.g., by using separation or washing techniques to eliminate unbound antibodies) and control antibody from the test antibody (e.g., by using species specific or isotype specific secondary antibodies or by specifically labelling the control antibody with a detectable label), the method can be used to determine that the test antibody reduces the binding of the control antibody to the COV-S antigen, indicating that the test
antibody recognizes substantially the same epitope as the control antibody (e.g., antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI- 58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, ADI-58131). The binding of the (labeled) control antibody in the presence of a completely irrelevant antibody (that does not bind CoV-S) can serve as the control high value. The control low value can be obtained by incubating the labeled control antibody with the same but unlabeled control antibody, where competition would occur and reduce binding of the labeled antibody. In a test assay, a significant reduction in labeled antibody reactivity in the presence of a test antibody is indicative of a test antibody that recognizes substantially the same epitope, i.e., one that competes with the labeled control antibody. For example, any test antibody that reduces the binding of any one of antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI- 58127, ADI-58128, ADI-58129, ADI-58130, ADI-58131, to both of SARS-CoV-S or SARS-CoV-2-S antigens by at least about 50%, such as at least about 60%, or more preferably at least about 70%
(e.g., about 65-100%), at any ratio of control antibody selected from the group consisting of ADI- 58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI- 58128, ADI-58129, ADI-58130, ADI-58131, test antibody between about 1:1 or 1:10 and about 1:100 is considered to be an antibody that binds to substantially the same epitope or determinant as any one of antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, ADI-58131, respectively. Preferably, such test antibody will reduce the binding of any one of antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI- 58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, ADI-58131, to at least one, preferably each, of the SARS-CoV-S or SARS-CoV-2-S antigens preferably at least about 50%, at least about 60%, at least about 80% or at least about 90% (e.g., about 95%) of the binding of any one of antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, ADI-58131, observed in the absence of the test antibody. These methods can be adapted to identify and/or evaluate antibodies that compete with other control antibodies.
[0354] A simple competition assay in which a test antibody is applied at saturating concentration to a surface onto which either SARS-CoV-S or SARS-CoV-2-S, or both, are immobilized also may be advantageously employed. The surface in the simple competition assay is preferably of a media suitable for OCTET® and/or PROTEON®. The binding of a control antibody (e.g., any one of antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, ADI-58131) to the CoV-S-coated surface is measured. This binding to the CoV-S-containing surface of the control antibody alone is compared with the binding of the control antibody in the presence of a test antibody.
A significant reduction in binding to the CoV-S-containing surface by the control antibody in the presence of a test antibody indicates that the test antibody recognizes substantially the same epitope as the control antibody such that the test antibody "competes" with the control antibody. Any test antibody that reduces the binding of control antibody (such as anyone of antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI- 58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, ADI-58131) to both of SARS-CoV-S and SARS-CoV-2-S antigens by at least about 20% or more, at least about 40%, at least about 50%, at least about 70%, or more, can be considered to be an antibody that binds to substantially the same epitope or determinant as the control antibody (e.g., any one of antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, ADI-58131). Preferably, such test antibody will reduce the binding of the control antibody (e.g., any one of antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, ADI-58131) to the CoV-S antigen by at least about 50% (e.g., at least about 60%, at least about 70%, or more). It will be appreciated that the order of control and test antibodies can be reversed; i.e. the control antibody can be first bound to the surface and then the test antibody is brought into contact with the surface thereafter in a competition assay. Preferably, the antibody having higher affinity for SARS-CoV-S and SARS-CoV-2-S is bound to the CoV-S-containing surface first, as it will be expected that the decrease in binding seen for the second antibody (assuming the antibodies are competing) will be of greater magnitude. Further examples of such assays are provided in, e.g., Saunal and Regenmortel, J. Immunol. Methods, 183:33-41 (1989), the disclosure of which is incorporated herein by reference.
[0355] Determination of whether an antibody, antigen-binding fragment thereof, or antibody derivative, e.g., an affinity-matured antibody or antigen binding fragment of any of the anti-CoV-S antibodies exemplified herein, binds within one of the epitope regions defined above can be carried out in ways known to the person skilled in the art. In another example of such mapping/characterization methods, an epitope region for an anti-CoV-S antibody may be determined by epitope "footprinting" using chemical modification of the exposed amines/carboxyls in the SARS- CoV-S and SARS-CoV-2-S protein. One specific example of such a foot-printing technique is the use of hydrogen-deuterium exchange detected by mass spectrometry (“HXMS”), wherein a hydrogen/deuterium exchange of receptor and ligand protein amide protons, binding, and back exchange occurs, wherein the backbone amide groups participating in protein binding are protected from back exchange and therefore will remain deuterated. Relevant regions can be identified at this point by peptic proteolysis, fast microbore high-performance liquid chromatography separation, and/or electrospray ionization mass spectrometry (See, e.g., Ehring H., Analytical Biochemistry, 267(2):252-259 (1999) and Engen, J. R. & Smith, D. L., Anal. Chem., 73:256A-265A (2001)).
Another example of a suitable epitope identification technique is nuclear magnetic resonance epitope mapping (“NMR”), where typically the position of the signals in two-dimensional NMR spectras of the free antigen and the antigen complexed with the antigen binding peptide, such as an antibody, are compared. The antigen typically is selectively isotopically labeled with 15N so that only signals corresponding to the antigen and no signals from the antigen binding peptide are seen in the NMR- spectrum. Antigen signals originating from amino acids involved in the interaction with the antigen binding peptide typically will shift position in the spectras of the complex compared to the spectras of the free antigen, and the amino acids involved in the binding can be identified that way. See, e.g., Ernst Sobering Res. Found. Workshop, (44):149-67 (2004); Huang etal., J. Mol. Biol., 281(1):61 -67 (1998); and Saito and Patterson, Methods, 9(3):516-24 (1996). Epitope mapping/characterization also can be performed using mass spectrometry (“MS”) methods {See, e.g., Downard, J. Mass Spectrom., 35(4):493-503 (2000) and Kiselar and Downard, Anal. Chem., 71(9):1792-801 (1999)).
[0356] Protease digestion techniques also can be useful in the context of epitope mapping and identification. Antigenic determinant-relevant regions/sequences can be determined by protease digestion, e.g. by using trypsin in a ratio of about 1:50 to SARS-CoV-S or SARS-CoV-2-S overnight (“o/n”) digestion at 37°C and pH 7-8, followed by mass spectrometry (“MS”) analysis for peptide identification. The peptides protected from trypsin cleavage by the anti-CoV-S antibody can subsequently be identified by comparison of samples subjected to trypsin digestion and samples incubated with antibody and then subjected to digestion by e.g. trypsin (thereby revealing a footprint for the antibody). Other enzymes like chymotrypsin or pepsin can be used in similar epitope characterization methods. Moreover, enzymatic digestion can provide a quick method for analyzing whether a potential antigenic determinant sequence is within a region of CoV-S in the context of a CoV-S-binding polypeptide. If the polypeptide is not surface exposed, it is most likely not relevant in terms of immunogenicity/antigenicity {See, e.g., Manca, Ann. 1st. Super. Sanita., 27(1): 15-9 (1991) for a discussion of similar techniques).
[0357] Site -directed mutagenesis is another technique useful for characterization of a binding epitope. For example, in "alanine-scanning" site -directed mutagenesis (also known as alanine scanning, alanine scanning mutagenesis, alanine scanning mutations, combinatorial alanine scanning, or creation of alanine point mutations, for example), each residue within a protein segment is replaced with an alanine residue (or another residue such as valine where alanine is present in the wild-type sequence) through such methodologies as direct peptide or protein synthesis, site-directed mutagenesis, the GENEART™ Mutagenesis Service (Thermo Fisher Scientific, Waltham, MA U.S.A.) or shotgun mutagenesis, for example. A series of single point mutants of the molecule is thereby generated using this technique; the number of mutants generated is equivalent to the number of residues in the molecule, each residue being replaced, one at a time, by a single alanine residue. Alanine is generally used to replace native (wild-type) residues because of its non-bulky, chemically
inert, methyl functional group that can mimic the secondary structure preferences that many other amino acids may possess. Subsequently, the effects replacing a native residue with an alanine has on binding affinity of an alanine scanning mutant and its binding partner can be measured using such methods as, but not limited to, SPR binding experiments. If a mutation leads to a significant reduction in binding affinity, it is most likely that the mutated residue is involved in binding. Monoclonal antibodies specific for structural epitopes (i.e., antibodies that do not bind the unfolded protein) can be used as a positive control for binding affinity experiments to verify that the alanine -replacement does not influence the overall tertiary structure of the protein (as changes to the overall fold of the protein may indirectly affect binding and thereby produce a false positive result). See, e.g., Clackson and Wells, Science, 267:383-386 (1995); Weiss et al, Proc. Natl. Acad. Sci. USA, 97(16):8950-8954 (2000); and Wells, Proc. Natl. Acad. Sci. USA, 93:1-6 (1996). Example 5 identifies the specific epitope or residues of CoV-S which specifically interact with the anti-CoV-S antibodies disclosed herein.
[0358] Electron microscopy can also be used for epitope "footprinting". For example, Wang et al. , Nature, 355:275-278 (1992) used coordinated application of cryoelectron microscopy, three- dimensional image reconstruction, and X-ray crystallography to determine the physical footprint of a Fab-fragment on the capsid surface of native cowpea mosaic virus.
[0359] Other forms of "label-free" assay for epitope evaluation include SPR (sold commercially as the BIACORE® system, GE Healthcare Life Sciences, Marlborough, MA) and reflectometric interference spectroscopy (“RifS”) (See, e.g., Fagerstam et al, Journal of Molecular Recognition, 3:208-14 (1990); Nice et al, J. Chromatogr., 646:159-168 (1993); Leipert et al, Angew. Chem. Int. Ed., 37:3308-3311 (1998); Kroger et al, Biosensors and Bioelectronics, 17:937-944 (2002)).
[0360] The expressions “framework region” or “FR” refer to one or more of the framework regions within the variable regions of the light and heavy chains of an antibody ( See Rabat et al. , Sequences of Proteins of Immunological Interest, 4th edition, Bethesda, MD: U.S. Dept of Health and Human Services, Public Health Service, National Institutes of Health (1987)). These expressions include those amino acid sequence regions interposed between the CDRs within the variable regions of the light and heavy chains of an antibody.
[0361] The term "Fc region" is used to define a C-terminal region of an immunoglobulin heavy chain. The "Fc region" may be a native sequence Fc region or a variant Fc region. Although the boundaries of the Fc region of an immunoglobulin heavy chain might vary, the human IgG heavy chain Fc region is usually defined to stretch from an amino acid residue at position Cys226, or from Pro230, to the carboxyl-terminus thereof. The numbering of the residues in the Fc region is that of the EU index as in Rabat. Rabat et al, Sequences of Proteins of Immunological Interest, 5th edition, Bethesda, MD: U.S. Dept of Health and Human Services, Public Health Service, National Institutes
of Health (1991). The Fc region of an immunoglobulin generally comprises two constant domains, Cm and CH3.
[0362] The terms "Fc receptor" and "FcR" describe a receptor that binds to the Fc region of an antibody. The preferred FcR is a native sequence human FcR. Moreover, a preferred FcR is one that binds an IgG antibody (a gamma receptor) and includes receptors of the FcyRI, FcyRII, and FcyRIII subclasses, including allelic variants and alternatively spliced forms of these receptors. FcyRII receptors include FcyRIIA (an "activating receptor") and FcyRIIB (an "inhibiting receptor"), which have similar amino acid sequences that differ primarily in the cytoplasmic domains thereof. FcRs are reviewed in Ravetch and Kinet, Ann. Rev. Immunol., 9:457-92 (1991); Capel et al, Immunomethods, 4:25-34 (1994); and de Haas et al, J. Lab. Clin. Med., 126:330-41 (1995). "FcR" also includes the neonatal receptor, FcRn, which is responsible for the transfer of maternal IgGs to the fetus (Guyer et al, J. Immunol., 117:587 (1976); and Kim et al, J. Immunol., 24:249 (1994)), and which primarily functions to modulate and/or extend the half-life of antibodies in circulation. To the extent that the disclosed anti-CoV-S antibodies are aglycosylated, as a result of the expression system and/or sequence, the subject antibodies are expected to bind FcRn receptors, but not to bind (or to minimally bind) Fey receptors.
[0363] A "functional Fc region" possesses at least one effector function of a native sequence Fc region. Exemplary "effector functions" include Clq binding; complement dependent cytotoxicity (“CDC”); Fc receptor binding; antibody-dependent cell-mediated cytotoxicity (“ADCC”); phagocytosis; down-regulation of cell surface receptors (e.g. B cell receptor (“BCR”)), etc. Such effector functions generally require the Fc region to be combined with a binding domain (e.g. an antibody variable domain) and can be assessed using various assays known in the art for evaluating such antibody effector functions.
[0364] A "native sequence Fc region" comprises an amino acid sequence identical to the amino acid sequence of an Fc region found in nature. A "variant Fc region" comprises an amino acid sequence that differs from that of a native sequence Fc region by virtue of at least one amino acid modification, yet retains at least one effector function of the native sequence Fc region. Preferably, the variant Fc region has at least one amino acid substitution compared to a native sequence Fc region or to the Fc region of a parent polypeptide, e.g. from about one to about ten amino acid substitutions, and preferably from about one to about five amino acid substitutions in a native sequence Fc region or in the Fc region of the parent polypeptide. The variant Fc region herein will preferably possess at least about 80% sequence identity with a native sequence Fc region and/or with an Fc region of a parent polypeptide, and most preferably at least about 90% sequence identity therewith, more preferably at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% sequence identity therewith.
[0365] In some embodiments, the Fc region of an antibody or antigen-binding antibody fragment of the present disclosure may bind to an Fc receptor (FcR). The FcR may be, but is not limited to, Fc gamma receptor (FcgR), FcgRI, FcgRIIA, FcgRIIBl, FcgRIIB2, FcgRIIIA, FcgRIIIB, Fc epsilon receptor (FceR), FceRI, FceRII, Fc alpha receptor (FcaR), FcaRI, Fc alpha/mu receptor (Fca/mR), or neonatal Fc receptor (FcRn). The Fc may be an IgM, IgD, IgG, IgE, or IgA isotype. An IgG isotype may be an IgGl, IgG2, IgG3, or IgG4.
[0366] Certain amino acid modifications in the Fc region are known to modulate Ab effector functions and properties, such as, but not limited to, antibody-dependent cellular cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP), complement dependent cytotoxicity (CDC), and half -life (Wang X. et ah, Protein Cell. 2018 Jan; 9(1): 63-73; DalFAcqua W. F. et ah, J Biol Chem. 2006 Aug 18;281(33):23514-24. Epub 2006 Jun 21; Monnet C. et al, Front Immunol. 2015 Feb 4;6:39. doi: 10.3389/fimmu.2015.00039. eCollection 2015). The mutation may be symmetrical or asymmetrical. In certain cases, antibodies with Fc regions that have asymmetrical mutation(s) (i.e., two Fc regions are not identical) may provide better functions such as ADCC (Fiu Z. et al. J Biol Chem. 2014 Feb 7; 289(6): 3571-3590).
[0367] Any of the antibody variable region sequences disclosed herein may be used in combination with a wild-type (WT) Fc or a variant Fc. In particular embodiments, an Fc selected from the Fc sequences described in Table 9 may be used. Any of the variable region sequences disclosed herein may be used in combination with any appropriate Fc including any of the Fc variants provided in Table 9 to form an antibody or an antigen-binding antibody fragment of the present disclosure. The lysine (K) at the C-terminus of each Fc may be present or absent.
[0368] An IgGl -type Fc optionally may comprise one or more amino acid substitutions. Such substitutions may include, for example, N297A, N297Q, D265A, F234A, F235A, C226S, C229S, P238S, E233P, F234V, G236-deleted, P238A, A327Q, A327G, P329A, K322A, F234F, F235E, P331S, T394D, A330F, P331S, F243F, R292P, Y300F, V305I, P396F, S239D, I332E, S298A, E333A, K334A, F234Y, F235Q, G236W, S239M, H268D, D270E, K326D, A330M, K334E, G236A, K326W, S239D, E333S, S267E, H268F, S324T, E345R, E430G, S440Y, M428F, N434S, F328F, M252Y, S254T, T256E, and/or any combination thereof (the residue numbering is according to the EU index as in Rabat) (DalFAcqua W. F. et ah, J Biol Chem. 2006 Aug 18;281(33):23514-24. Epub 2006 Jun 21; Wang X. et al, Protein Cell. 2018 Jan; 9(1): 63-73), or for example, N434A, Q438R, S440E, F432D, N434F, and/or any combination thereof (the residue numbering according to EU numbering). The Fc region may further comprise one or more additional amino acid substitutions. Such substitutions may include but are not limited to A330F, F234F, F235E, P3318, and/or any combination thereof (the residue numbering is according to the EU index as in Rabat). Specific exemplary substitution combinations for an IgGl -type Fc include, but not limited to: M252Y, S254T, and T256E (“YTE” variant); M428F and N434A (“FA” variant), M428F and N434S (“LS” variant);
M428L, N434A, Q438R, and S440E (“LA-RE” variant); L432D and N434L (“DEL” variant); and L234A, L235A, L432D, and N434L (“LALA-DEL” variant) (the residue numbering is according to the EU index as in Rabat). In particular embodiments, an IgGl-type Fc variant may comprise the amino acid sequence of SEQ ID NOS: 411, 412, 413, 414, 415, 416, or 417. In one embodiment, the Fc variant is an LA variant and comprises the amino acid sequence of SEQ ID NO: 413.
[0369] When the Ab is an IgG2, the Fc region optionally may comprise one or more amino acid substitutions. Such substitutions may include but are not limited to P238S, V234A, G237A, H268A, H268Q, H268E, V309L, N297A, N297Q, A330S, P331S, C232S, C233S, M252Y, S254T, T256E, and/or any combination thereof (the residue numbering is according to the EU index as in Rabat). The Fc region optionally may further comprise one or more additional amino acid substitutions. Such substitutions may include but are not limited to M252Y, S254T, T256E, and/or any combination thereof (the residue numbering is according to the EU index as in Rabat).
[0370] An IgG3-type Fc region optionally may comprise one or more amino acid substitutions. Such substitutions may include but are not limited to E235Y (the residue numbering is according to the EU index as in Rabat).
[0371] An IgG4-type Fc region optionally may comprise one or more amino acid substitutions. Such substitutions may include but are not limited to, E233P, F234V, L235A, G237A, E318A, S228P, L236E, S241P, L248E, T394D, M252Y, S254T, T256E, N297A, N297Q, and/or any combination thereof (the residue numbering is according to the EU index as in Rabat). The substitution may be, for example, S228P (the residue numbering is according to the EU index as in Rabat).
[0372] In some cases, the glycan of the hum an -like Fc region may be engineered to modify the effector function (for example, see Li T. et al, Proc Natl Acad Sci USA. 2017 Mar 28; 114(13):3485- 3490. doi: 10.1073/pnas.l702173114. Epub 2017 Mar 13).
[0373] An “isolated” antibody, as used herein, refers to an antibody that is substantially free of other antibodies having different antigenic specificities. In some embodiments, an isolated antibody is substantially free of other unintended cellular material and/or chemicals.
[0374] As used herein, “specific binding” or “specifically binds” means that the interaction of the antibody, or antigen-binding portion thereof, with an antigen is dependent upon the presence of a particular structure (e.g., antigenic determinant or epitope). For example, the antibody, or antigen binding portion thereof, binds to a specific protein, rather than proteins generally. In some embodiments, an antibody, or antigen-binding portion thereof, specifically binds a target, e.g., SARS- CoV-S and/or SARS-CoV-S-2. In some embodoiments, an antibody, or antigen-binding portion thereof, specifically binds to more than one coronavirus spike protein, e.g., the spike protein of SARS-CoV-S and the spike protein of SARS-CoV-2-S, for example. In some embodiments, the antibody, or antigen-binding portion thereof, specifically binds to two different, but related, antigens,
e.g., the spike protein of SARS-CoVl-S and the spike protein of SARS-CoV2-S, e.g., via a conserved epitope.
B. Anti-CoV-S Antibodies and Binding Fragments Thereof Having Binding Activity for CoV-S [0375] CoV-S refers to the S protein of a coronavirus which is expressed on the surface of virions as a structural protein. As mentioned previously, the S protein plays an essential role for coronaviruses in binding to receptors on the host cell and determines host tropism (Zhu Z. et al. , Infect Genet Evol. 2018 Jul;61 : 183-184). SARS-CoV and SARS-CoV-2 bind to angiotensin-converting enzyme 2 (ACE2) of the host cell via the S protein’s receptor-binding domains (RBDs) and uses ACE2 as a receptor to enter the host cells (Ge X.Y. et al, Nature. 2013 Nov 28;503(7477):535-8. doi:
10.1038/nature 12711. Epub 2013 Oct 30.; Hoffmann M. et al., Cell. 2020 Mar 4. pii: S0092- 8674(20)30229-4). SARS-CoV can also use CD209L (also known as L-SIGN) as an alternative receptor (Jeffers S. A. et al, Proc Natl Acad Sci USA. 2004 Nov 2;101(44):15748-53. Epub 2004 Oct 20). MERS-CoV binds dipeptidyl peptidase 4 (“DPP4”, also known as CD26) of the host cells via a different RBD of the S protein. Cell entry of coronaviruses depends on not only binding of the S protein to a host cell receptor but often also priming of the S protein by host cell proteases, and recently SARS-CoV-2 was found to use the serine protease TMPRSS2 for S protein priming and then ACE2 for entry (Wu A. et al, Cell Host Microbe. 2020 Mar ll;27(3):325-328; Hoffmann M. et al, Cell. 2020 Mar 4. pii: S0092-8674(20)30229-4).
[0376] The S protein of SARS-CoV is referred to as SARS-CoV-S and may for example comprise the amino acid sequence of SEQ ID NO: 401 (1288 amino acids). The S protein of SARS-CoV-2 is referred to as SARS-CoV -2-S and may for example comprise the amino acid sequence of SEQ ID NO: 403 (1273 amino acids).
[0377] The present disclosure provides exemplary antibodies and antigen-binding antibody fragments that specifically bind to CoV, wherein at least some of these antibodies and antigen-binding antibody fragments specifically bind to SARS-CoV -2-S and/or SARS-CoV-2-S. Due to the sequence similarity among different CoV species, such antibodies or antigen-binding antibody fragments of the present disclosure may also cross react with the S protein of other CoV species.
[0378] The exemplary S proteins of CoV that the antibodies or antigen-binding antibody fragments of the present disclosure may specifically bind include by way of example, Bat SARS CoV (GenBank Accession No. FJ211859), SARS CoV (GenBank Accession No. FJ211860), BtSARS.HKU3.1 (GenBank Accession No. DQ022305), BtSARS.HKU3.2 (GenBank Accession No. DQ084199), BtSARS.HKU3.3 (GenBank Accession No. DQ084200), BtSARS.Rml (GenBank Accession No. DQ412043), BtCoV.279.2005 (GenBank Accession No. DQ648857), BtSARS.Rfl (GenBank Accession No. DQ412042), BtCoV.273.2005 (GenBank Accession No. DQ648856), BtSARS.Rp3 (GenBank Accession No. DQ071615), SARS CoV.A022 (GenBank Accession No. AY686863),
SARSCoV.CUHK-W 1 (GenBank Accession No. AY278554), SARSCoV.GDOl (GenBank Accession No. AY278489), SARSCoV.HC.SZ.61.03 (GenBank Accession No. AY515512), SARSC0V.SZI6 (GenBank Accession No. AY304488), SARSCoV.Urbani (GenBank Accession No. AY278741), SARSCoV.civetOlO (GenBank Accession No. AY572035), or SARSCoV.MA.15 (GenBank Accession No. DQ497008), Rs SHC014 (GenBank® Accession No. KC881005), Rs3367 (GenBank® Accession No. KC881006), WiVl S (GenBank® Accession No. KC881007).
[0379] In some embodiments, the antibodies and antigen-binding antibody fragments provided herein may also bind to and neutralize existing bat CoV or pre-emergent bat CoVs. Antibodies and antigen binding antibody fragments with such binding and/or neutralization abilities would be particularly useful in a future pandemic that may be caused by a spillover from an animal reservoir, like a bat. In fact, ADI-55688, ADI-55689, ADI-55993, ADI-5600, ADI-56046, ADI-55690, ADI-56010, and ADI-55951 were shown to neutralize authentic bat coronavirus, WIV1 (see Figure 3E of Wee A. et al, Science. 2020 Jun 15;eabc7424. doi: 10.1126/science.abc7424).
[0380] Alternatively, the S proteins of CoV to which the antibodies or antigen-binding antibody fragments of the present disclosure may specifically bind to and neutralize pre-emergent coronaviruses from other species, e.g., bats.
[0381] Still alternatively, the S proteins of CoV to which the antibodies or antigen-binding antibody fragments of the present disclosure may specifically bind to may include, for example, Middle East respiratory syndrome coronavirus isolate Riyadh_2_2012 (GenBank Accession No. KF600652.1), Middle East respiratory syndrome coronavirus isolate Al-Hasa_18_2013 (GenBank Accession No. KF600651.1), Middle East respiratory syndrome coronavirus isolate Al-Hasa_17_2013 (GenBank Accession No. KF600647.1), Middle East respiratory syndrome coronavirus isolate Al-Hasa_15_2013 (GenBank Accession No. KF600645.1), Middle East respiratory syndrome coronavirus isolate Al- Hasa_16_2013 (GenBank Accession No. KF600644.1), Middle East respiratory syndrome coronavirus isolate Al-Hasa_21_2013 (GenBank Accession No. KF600634), Middle East respiratory syndrome coronavirus isolate Al-Hasa_19_2013 (GenBank Accession No. KF600632), Middle East respiratory syndrome coronavirus isolate Buraidah_l_2013 (GenBank Accession No. KF600630.1), Middle East respiratory syndrome coronavirus isolate Hafr-Al-Batin_l_2013 (GenBank Accession No. KF600628.1), Middle East respiratory syndrome coronavirus isolate Al-Hasa_12_2013 (GenBank Accession No. KF600627.1), Middle East respiratory syndrome coronavirus isolate Bisha_l_2012 (GenBank Accession No. KF600620.1), Middle East respiratory syndrome coronavirus isolate Riyadh_3_2013 (GenBank Accession No. KF600613.1), Middle East respiratory syndrome coronavirus isolate Riyadh_l_2012 (GenBank Accession No. KF600612.1), Middle East respiratory syndrome coronavirus isolate Al-Hasa_3_2013 (GenBank Accession No. KF186565.1), Middle East respiratory syndrome coronavirus isolate Al-Hasa_l_2013 (GenBank Accession No. KF186567.1), Middle East respiratory syndrome coronavirus isolate Al-Hasa_2_2013 (GenBank Accession No.
KF186566.1), Middle East respiratory syndrome coronavirus isolate Al-Hasa_4_2013 (GenBank Accession No. KF186564.1), Middle East respiratory syndrome coronavirus (GenBank Accession No. KF192507.1), Betacoronavirus England 1-Nl (GenBank Accession No. NC_019843), MERS- CoV_SA-Nl (GenBank Accession No. KC667074), following isolates of Middle East Respiratory Syndrome Coronavirus (GenBank Accession No: KF600656.1, GenBank Accession No: KF600655.1, GenBank Accession No: KF600654.1, GenBank Accession No: KF600649.1, GenBank Accession No: KF600648.1, GenBank Accession No: KF600646.1, GenBank Accession No: KF600643.1, GenBank Accession No: KF600642.1, GenBank Accession No: KF600640.1, GenBank Accession No: KF600639.1, GenBank Accession No: KF600638.1, GenBank Accession No: KF600637.1, GenBank Accession No: KF600636.1, GenBank Accession No: KF600635.1, GenBank Accession No: KF600631.1, GenBank Accession No: KF600626.1, GenBank Accession No: KF600625.1, GenBank Accession No: KF600624.1, GenBank Accession No: KF600623.1, GenBank Accession No: KF600622.1, GenBank Accession No: KF600621.1, GenBank Accession No: KF600619.1, GenBank Accession No: KF600618.1, GenBank Accession No: KF600616.1, GenBank Accession No: KF600615.1, GenBank Accession No: KF600614.1, GenBank Accession No: KF600641.1, GenBank Accession No: KF600633.1, GenBank Accession No: KF600629.1, GenBank Accession No: KF600617.1), Coronavirus Neoromicia/PML-PHEl/RSA/2011 GenBank Accession: KC869678.2, Bat Coronavirus Taper/CII_KSA_287/Bisha/Saudi Arabia/GenBank Accession No: KF493885.1, Bat coronavirus Rhhar/CII_KSA_003/Bisha/Saudi Arabia/2013 GenBank Accession No: KF493888.1, Bat coronavirus Pikuh/CII_KSA_001/Riyadh/Saudi Arabia/2013 GenBank Accession No: KF493887.1, Bat coronavirus Rhhar/CII_KSA_002/Bisha/Saudi Arabia/2013 GenBank Accession No: KF493886.1, Bat Coronavirus Rhhar/CII_KSA_004/Bisha/Saudi Arabia/2013 GenBank Accession No: KF493884.1, BtCoV.HKU4.2 (GenBank Accession No. EF065506), BtCoV.HKU4.1 (GenBank Accession No. NC_009019), BtCoV.HKU4.3 (GenBank Accession No. EF065507), BtCoV.HKU4.4 (GenBank Accession No. EF065508), BtCoV 133.2005 (GenBank Accession No. NC 008315), BtCoV.HKU5.5 (GenBank Accession No. EF065512); BtCoV.HKU5.1 (GenBank Accession No. NC_009020), BtCoV.HKU5.2 (GenBank Accession No. EF065510), BtCoV.HKU5.3 (GenBank Accession No. EF065511), human betacoronavirus 2c Jordan- N3/2012 (GenBank Accession No. KC776174.1; human betacoronavirus 2c EMC/2012 (GenBank Accession No. JX869059.2), Pipistrellus bat coronavirus HKU5 isolates (GenBank Accession No:KC522089.1, GenBank Accession No:KC522088.1, GenBank Accession No:KC522087.1, GenBank Accession No:KC522086.1, GenBank Accession No:KC522085.1, GenBank Accession No: KC522084.1, GenBank Accession No:KC522083.1, GenBank Accession No:KC522082.1, GenBank Accession No:KC522081.1, GenBank Accession No:KC522080.1, GenBank Accession No:KC522079.1, GenBank Accession No:KC522078.1, GenBank Accession No: KC522077.1, GenBank Accession No:KC522076.1, GenBank Accession No:KC522075.1, GenBank Accession
No:KC522104.1, GenBank Accession No:KC522104.1, GenBank Accession No:KC522103.1, GenBank Accession No:KC522102.1, GenBank Accession No: KC522101.1, GenBank Accession No:KC522100.1, GenBank Accession No:KC522099.1, GenBank Accession No:KC522098.1, GenBank Accession No:KC522097.1, GenBank Accession No:KC522096.1, GenBank Accession No:KC522095.1, GenBank Accession No: KC522094.1, GenBank Accession No:KC522093.1, GenBank Accession No:KC522092.1, GenBank Accession No:KC522091.1, GenBank Accession No:KC522090.1, GenBank Accession No:KC522119.1 GenBank Accession No:KC522118.1 GenBank Accession No: KC522117.1 GenBank Accession No:KC522116.1 GenBank Accession No:KC522115.1 GenBank Accession No:KC522114.1 GenBank Accession No:KC522113.1 GenBank Accession No:KC522112.1 GenBank Accession No:KC522111.1 GenBank Accession No: KC522110.1 GenBank Accession No:KC522109.1 GenBank Accession No:KC522108.1, GenBank Accession No:KC522107.1, GenBank Accession No:KC522106.1, GenBank Accession No:KC522105.1) Pipistrellus bat coronavirus HKU4 isolates (GenBank Accession No:KC522048.1, GenBank Accession No:KC522047.1, GenBank Accession No:KC522046.1, GenBank Accession No:KC522045.1, GenBank Accession No: KC522044.1, GenBank Accession No:KC522043.1, GenBank Accession No:KC522042.1, GenBank Accession No:KC522041.1, GenBank Accession No:KC522040.1 GenBank Accession No:KC522039.1, GenBank Accession No:KC522038.1, GenBank Accession No:KC522037.1, GenBank Accession No:KC522036.1, GenBank Accession No:KC522048.1 GenBank Accession No:KC522047.1 GenBank Accession No:KC522046.1 GenBank Accession No:KC522045.1 GenBank Accession No:KC522044.1 GenBank Accession No:KC522043.1 GenBank Accession No:KC522042.1 GenBank Accession No:KC522041.1 GenBank Accession No:KC522040.1, GenBank Accession No:KC522039.1 GenBank Accession No:KC522038.1 GenBank Accession No:KC522037.1 GenBank Accession No:KC522036.1, GenBank Accession No:KC522061.1 GenBank Accession No:KC522060.1 GenBank Accession No:KC522059.1 GenBank Accession No:KC522058.1 GenBank Accession No:KC522057.1 GenBank Accession No:KC522056.1 GenBank Accession No:KC522055.1 GenBank Accession No:KC522054.1 GenBank Accession No:KC522053.1 GenBank Accession No:KC522052.1 GenBank Accession No:KC522051.1 GenBank Accession No:KC522050.1 GenBank Accession No:KC522049.1 GenBank Accession No:KC522074.1, GenBank Accession No:KC522073.1 GenBank Accession No:KC522072.1 GenBank Accession No:KC522071.1 GenBank Accession No:KC522070.1 GenBank Accession No:KC522069.1 GenBank Accession No:KC522068.1 GenBank Accession No:KC522067.1, GenBank Accession No:KC522066.1 GenBank Accession No:KC522065.1 GenBank Accession No:KC522064.1, GenBank Accession No:KC522063.1, or GenBank Accession No:KC522062.1.
[0382] Alternatively, the S proteins of CoV to which the antibodies or antigen-binding antibody fragments of the present disclosure may specifically bind may include for example,
FCov.FIPV.79.1146.VR.2202 (GenBank Accession No. NV_007025), transmissible gastroenteritis virus (TGEV) (GenBank Accession No. NC_002306; GenBank Accession No. Q811789.2; GenBank Accession No. DQ811786.2; GenBank Accession No. DQ811788.1; GenBank Accession No. DQ811785.1; GenBank Accession No. X52157.1; GenBank Accession No. AJ011482.1; GenBank Accession No. KC962433.1; GenBank Accession No. AJ271965.2; GenBank Accession No. JQ693060.1; GenBank Accession No. KC609371.1; GenBank Accession No. JQ693060.1; GenBank Accession No. JQ693059.1; GenBank Accession No. JQ693058.1; GenBank Accession No. JQ693057.1; GenBank Accession No. JQ693052.1; GenBank Accession No. JQ693051.1; GenBank Accession No. JQ693050.1), or porcine reproductive and respiratory syndrome virus (PRRSV) (GenBank Accession No. NC_001961.1; GenBank Accession No. DQ811787).
[0383] Alternatively, the S proteins of CoV to which the antibodies or antigen-binding antibody fragments of the present disclosure may specifically bind may include, for example,
BtCoV.1 A.AFCD62 (GenBank Accession No. NC_010437), BtCoV.lB.AFCD307 (GenBank Accession No. NC_010436), BtCov.FlKU8.AFCD77 (GenBank Accession No. NC_010438), BtCoV.512.2005 (GenBank Accession No. DQ648858), porcine epidemic diarrhea virus PEDV.CV777 (GenBank Accession No. NC_003436, GenBank Accession No. DQ355224.1, GenBank Accession No. DQ355223.1, GenBank Accession No. DQ355221.1, GenBank Accession No. JN601062.1, GenBank Accession No. N601061.1, GenBank Accession No. JN601060.1, GenBank Accession No. JN601059.1, GenBank Accession No. JN601058.1, GenBank Accession No. JN601057.1, GenBank Accession No. JN601056.1, GenBank Accession No. JN601055.1, GenBank Accession No. JN601054.1, GenBank Accession No. JN601053.1, GenBank Accession No. JN601052.1, GenBank Accession No. JN400902.1, GenBank Accession No. JN547395.1, GenBank Accession No. FJ687473.1, GenBank Accession No. FJ687472.1, GenBank Accession No. FJ687471.1, GenBank Accession No. FJ687470.1, GenBank Accession No. FJ687469.1, GenBank Accession No. FJ687468.1, GenBank Accession No. FJ687467.1, GenBank Accession No. FJ687466.1, GenBank Accession No. FJ687465.1, GenBank Accession No. FJ687464.1, GenBank Accession No. FJ687463.1, GenBank Accession No. FJ687462.1, GenBank Accession No. FJ687461.1, GenBank Accession No. FJ687460.1, GenBank Accession No. FJ687459.1, GenBank Accession No. FJ687458.1, GenBank Accession No. FJ687457.1, GenBank Accession No. FJ687456.1, GenBank Accession No. FJ687455.1, GenBank Accession No. FJ687454.1, GenBank Accession No. FJ687453 GenBank Accession No. FJ687452.1, GenBank Accession No. FJ687451.1, GenBank Accession No. FJ687450.1, GenBank Accession No. FJ687449.1, GenBank Accession No. AF500215.1, GenBank Accession No. KF476061.1, GenBank Accession No. KF476060.1, GenBank Accession No. KF476059.1, GenBank Accession No. KF476058.1, GenBank Accession No. KF476057.1, GenBank Accession No. KF476056.1, GenBank Accession No. KF476055.1, GenBank Accession No. KF476054.1, GenBank Accession No. KF476053.1, GenBank Accession No.
KF476052.1, GenBank Accession No. KF476051.1, GenBank Accession No. KF476050.1, GenBank Accession No. KF476049.1, GenBank Accession No. KF476048.1, GenBank Accession No. KF177258.1, GenBank Accession No. KF177257.1, GenBank Accession No. KF177256.1, GenBank Accession No. KF177255.1), HCoV.229E (GenBank Accession No. NC_002645), HCoV.NL63.Amsterdam.I (GenBank Accession No. NC_005831), BtCoV.FIKU2.FIK.298.2006 (GenBank Accession No. EF203066), BtCoV.FIKU2.FIK.33.2006 (GenBank Accession No. EF203067), BtCoV.HKU2.HK.46.2006 (GenBank Accession No. EF203065), or BtCoV.FIKU2.GD.430.2006 (GenBank Accession No. EF203064).
[0384] Alternatively, the S proteins of CoV to which the antibodies or antigen-binding antibody fragments of the present disclosure may specifically bind may include, for example, FtCoV.FlKUl.C.N5 (GenBank Accession No. DQ339101), MFFV.A59 (GenBank Accession No. NC 001846), PHEV.VW572 (GenBank Accession No. NC 007732), HCoV.OC43.ATCC.VR.759 (GenBank Accession No. NC_005147), or bovine enteric coronavirus (BCoV.ENT) (GenBank Accession No. NC_003045).
[0385] Alternatively, the S proteins of CoV to which the antibodies or antigen-binding antibody fragments of the present disclosure may specifically bind may include, for example, BtCoV.HKU9.2 (GenBank Accession No. EF065514), BtCoV.HKU9.1 (GenBank Accession No. NC_009021), BtCoV.HkU9.3 (GenBank Accession No. EF065515), or BtCoV.HKU9.4 (GenBank Accession No. EF065516).
[0386] In some instances, an anti-CoV-S antibody or antigen-binding fragment thereof according to the disclosure binds to CoV-S (e.g., SARS-CoV-S and/or SARS-CoV-2-S, and/or any of the CoV S proteins listed above) with a dissociation constant (KD) of (i) 100 nM or lower; (ii) about 10 nM or lower; (iii) about 1 nM or lower; (iv) about 100 pM or lower; (v) about 10 pM or lower; (vi) about 1 pM or lower; or (vii) about 0.1 pM or lower.
[0387] The present disclosure provides exemplary antibodies or antigen-binding fragments thereof that bind CoV-S, including human CoV-S, which optionally may be affinity-matured. Other antibodies or antigen-binding fragments thereof that bind CoV-S, including those having different CDRs, and epitopic specificity may be obtained using the disclosure of the present specification, and using methods that are generally known in the art. Such antibodies and antigen-binding fragments thereof antagonize the biological effects of CoV-S in vivo and therefore are useful in treating or preventing COV-S-related conditions including, particularly coronavirus infection. In preferred embodiments, the antibody or antigen-binding fragment thereof according to the disclosure comprises one or more CDRs, a VL chain and/or VH chain of the anti-CoV-S antibodies and antigen-binding fragments thereof described herein.
[0388] In some embodiments, an anti-CoV-S antibody or antigen-binding fragment thereof according to the disclosure will interfere with, block, reduce, or modulate the interaction between COV-S and its
receptor(s) (e.g., ACE2, CD209L, L-SIGN, DPP4, or CD26) on host cells or a S protein-priming protein on host cells (e.g., TMPRSS2). If binding of the S protein to its receptor is blocked or reduced, CoV virions may be prohibited from entering the cells, i.e., infection to further cells is prevented.
Also, if the S protein is prevented from binding to a S protein-priming protein, the S protein would not be activated and therefore the host cell entry via the receptor may be reduced, i.e., infection to further cells is prevented.
[0389] In some instance, an anti-CoV-S antibody or antigen-binding fragment thereof according to the disclosure is “neutralizing”, e.g., it substantially or totally prevents the specific interaction of CoV-S with the host receptors or priming protein. As a result, CoV virions may be substantially or totally cleared by immune cells of the host, such as phagocytes via, for example, Fc receptor mediated phagocytosis or mere phagocytosis due to increased time of virions outside the cells. In some embodiments, the antibody or antigen-binding fragment thereof neutralizes CoV-S, e.g., by remaining bound to CoV-S in a location and/or manner that prevents CoV-S from specifically binding to its receptor or priming protein on host cells. As a result, CoV virions may be substantially or totally prevented from entering the cells, i.e. infection to further cells is prevented. In certain embodiments, an anti-CoV-S antibody or antigen-binding fragment thereof according to the disclosure neutralizes CoV (e.g., SARS-CoV and/or SARS-CoV-2) at an IC50 of about 100 nM or lower, of about 50 nM or lower, of about 20 nM or lower, of about 10 nM or lower, of about 5 nM or lower, of about 2 nM or lower, of about 1 nM or lower, of about 500 pM or lower, of about 200 pM or lower, of about 100 pM or lower, of about 50 pM or lower, of about 20 pM or lower, of about 10 pM or lower, of about 5 pM or lower, of about 2 pM or lower, or of about 1 pM or lower, or at an IC50 of about 5 μg/mL or lower, of about 4.5 μg/mL, of about 4 μg/mL, of about 3.5 μg/mL, of about 3 μg/mL, of about 2.5 μg/mL, of about 2 μg/mL or lower, of about 1.5 μg/mL or lower, of about 1 μg/mL or lower, of about about 500 ng/mL or lower, about 500 ng/mL or lower, of about 400 ng/mL or lower, of about 200 ng/mL or lower, of about 100 ng/mL or lower, of about 50 ng/mL or lower, at about 20 ng/mL or lower, at about 10 ng/mL or lower, at about 5 ng/mL or lower, at about 2 ng/mL or lower, or at about 1 ng/mL or lower, or about 0.6 ng/mL or lower, or of about 0.1 ng/mL to about 5 μg/mL, of about 0.5 ng/mL to about 4.5 μg/mL, of about 0.5 ng/mL to about 4 μg/mL, of about 0.5 ng/mL to about 3.5 μg/mL, of about 0.5 ng/mL to about 3 μg/mL, of about 0.5 ng/mL to about 2.5 μg/mL, of about 0.5 ng/mL to about 2 μg/mL, of about 1 ng/mL to about 2 μg/mL, of about 1 ng/mL to about 1.5 μg/mL, of about 100 ng/mL to about 1.1 μg/mL, of about 100 ng/mL to about 1 μg/mL, of about 100 ng/mL to about 500 ng/mL, of about 400 ng/mL to about 1.1 μg/mL, of about 300 ng/mL to about 1.1 μg/mL, of about 250 ng/mL to about 1.1 μg/mL, of about 200 ng/mL to about 1.1 μg/mL, in vitro, as measured by any of the neutralization assays described in Examples herein. In another embodiment, the isolated antibody or antigen-binding antibody fragment may neutralize SARS-CoV and/or SARS- CoV-2 at an IC90 of about 10 μg/mL or less, an IC90 of about 1 to about 10 μg/mL, an IC90 of about
2 to about 10 mg/mL, an IC90 of about 3 to about 10 mg/mL, an IC90 of about 5 to 10 mg/mL, an IC90 of about 7 to 10 mg/mL, an IC90 of about 8 to 10 mg/mL, an IC90 of about 9 to 10 mg/mL, an IC90 of about 10 mg/mL, 9 mg/mL, 8 mg/mL, 7, mg/mL, 6 mg/mL, or 5 mg/mL.
[0390] In some instances, an anti-CoV-S antibody or antigen-binding fragment thereof according to the disclosure or cocktail thereof, when administered to a coronavirus infected host or one susceptible to coronavirus infection such as a health care worker may promote a neutralization response in the host against the coronavirus which is sufficient to permit the host to be able to mount an effective cell-mediated immune response against the virus, e.g., T cell-mediated or cytokine-mediated immune response against the coronavirus and/or to be more responsive to other treatment methods such as drugs, antivirals or other biologies.
[0391] As mentioned, the anti-CoV-S antibodies or antigen-binding fragments thereof according to the disclosure have a variety of uses. For example, the subject antibodies and fragments can be useful in prophylactic or therapeutic applications, as well as diagnostically in binding assays. The subject anti-CoV-S antibodies or antigen-binding fragments thereof are useful for affinity purification of CoV-S, in particular human CoV-S or its ligands and in screening assays to identify other antagonists of CoV-S activity. Some of the antibodies or antigen-binding fragments thereof are useful for inhibiting binding of CoV-S to its receptor(s) (e.g., ACE2, CD209L, L-SIGN, DPP4, or CD26) on host cells or a S protein-priming protein on host cells (e.g., TMPRSS2) or inhibiting COV-S-mediated activities and/or biological effects.
[0392] As used herein, the term “one or more biological effects associated with COV-S refers to any biological effect mediated, induced, or otherwise attributable to COV-S, e.g., binding properties, functional properties, and other properties of biological significance. Non-limiting exemplary biological effects of COV-S include COV-S binding to its receptor(s) (e.g., ACE2, CD209L, L-SIGN, DPP4, or CD26) on host cells or a S protein-priming protein on host cells (e.g., TMPRSS2), activation of host cells for allowing virus entry, activation of immune cells as a result of the entry of CoV into the cell, e.g., via presentation of CoV antigen(s) on the host cells’ MHC molecule, and resulting inflammation. The subject anti-CoV-S antibodies are capable of inhibiting one, a combination of, or all of these exemplary CoV-S biological activities. For example, the anti-CoV-S antibodies and antigen-binding fragments thereof provided herein may neutralize CoV virions or reduce the infectivity of CoV virions.
[0393] The antibody or antigen-binding fragment thereof according to the disclosure can be used in a variety of therapeutic applications. For example, in some embodiments the anti-CoV-S antibody or antigen-binding fragment thereof are useful for treating conditions associated with CoV-S, such as, but not limited to, symptoms associated with CoV infection. The CoV may be any CoV, including SARS-CoV, SARS-CoV-2, MERS-CoV, HCoV-HKUl, HCoV-OC43, HCoV-229E, and HCoV- NL63, and also may be any of the CoV species listed above herein.
[0394] Specific examples of CoV infection-associated symptoms are fever, cough, dry cough, shortness of breath or difficulty of breath, fatigue, aches, runny nose, congestion, sore throat, conjunctivitis, chest pain, headache, muscle ache, chills, loss of smell, and loss of taste, and gastrointestinal symptoms including diarrhea. Complications and/or diseases/disorders associated with coronavirus infection may include, for example, bronchitis, pneumonia, respiratory failure, acute respiratory failure, organ failure, multi-organ system failure, pediatric inflammatory multisystem syndrome, acute respiratory distress syndrome (a severe lung condition that causes low oxygen in the blood and organs), blood clots, cardiac conditions, myocardial injury, myocarditis, heart failure, cardiac arrest, acute myocardial infarction, dysrhythmias, venous thromboembolism, post-intensive care syndrome, shock, anaphylactic shock, cytokine release syndrome, septic shock, disseminated intravascular coagulation, ischemic stroke, intracerebral hemorrhage, microangiopathic thrombosis, psychosis, seizure, nonconvulsive status epilepticus, traumatic brain injury, stroke, anoxic brain injury, encephalitis, posterior reversible leukoencephalopathy, necrotizing encephalopathy, post- infectious encephalitis, autoimmune mediated encephalitis, acute disseminated encephalomyelitis, acute kidney injury, acute liver injury, pancreatic injury, immune thrombocytopenia, subacute thyroiditis, gastrointestinal complications, aspergillosis, increased susceptibility to infection with another virus or bacteria, and/or pregnancy-related complications. Certain diseases and conditions, such as high blood pressure, type 1 diabetes, liver disease, overweight, chronic lung diseases including cystic fibrosis, pulmonary fibrosis, and asthma, compromised immune system due to transplant, use of an immunosuppressant, or HIV infection, and brain and nervous sustem condition, may increase the risk of CoV infection-associated complications and diseases.
[0395] The subject anti-CoV-S antibodies and antigen-binding fragments thereof may be used alone or in association with other active agents or drugs, including other biologies, to treat any subject in which blocking, inhibiting, or neutralizing the in vivo effect of CoV-S or blocking or inhibiting the interaction of CoV-S and its receptor(s) (e.g., ACE2, CD209L, L-SIGN, DPP4, or CD26) on host cells or a S protein-priming protein on host cells (e.g., TMPRSS2), is therapeutically desirable. In some embodiment, the subject anti-CoV-S antibody and antigen-binding fragment thereof, e.g., ADI- 58125, may be used in combination with a second antibody, or antigen-binding fragment thereof, wherein the second antibody, or antigen-binding fragment thereof, is selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, ADI-58131, or a combination thereof. In some embodiment, the second antibody, or antigen-binding fragment thereof, is ADI-58122. In one embodiment, the second antibody, or antigen-binding fragment thereof, is ADI-58127. In one embodiment, the second antibody, or antigen-binding fragment thereof, is ADI-58129. In one embodiment, the second antibody, or antigen-binding fragment thereof, is ADI-58131.
[0396] Exemplary anti-CoV antibodies and antigen-binding fragments thereof according to the disclosure, and the specific CDRs thereof are identified in this section.
[0397] The anti-CoV-S antibodies and antigen-binding fragments thereof comprising the disclosure have binding affinity for CoV-S, such as SARS-CoV-S or SARS-CoV-S2. Some antibodies of the present disclosure bind to SARS-CoV-S or SARS-CoV-S2 with a similar KD (M), while some antibodies of the present disclosure bind to SARS-CoV-S with a lower KD (M) ( /'. e. , higher affinity) than to SARS-CoV-S2, and some antibodies of the present disclosure bind to SARS-CoV-S-2 with a lower KD (M) (i.e., higher affinity) than to SARS-CoV-S.
C. Anti-CoV-S Antibody Polypeptide Sequences and Nucleic Acid Sequences Encoding Thereof Antibodies disclosed herein
[0398] Anti-CoV-S antibodies, and antigen-binding fragments thereof, specifically provided herein include: antibodies ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI- 58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, ADI-58131, and antigen-binding fragments thereof. Any Fc variant including but not limited to those specifically disclosed in Table 9 may be used in combination with any of the variable sequences disclosed herein. In some embodiments, the Fc variant is an FA variant and comprises the amino acid sequence of SEQ ID NO: 413. In one embodiment, the antibody ADI-58125 comprises an Fc variant of SEQ ID NO:413.
[0399] Tables 1-8 show the SEQ ID NOs assigned to individual amino acid sequences of the HC, VH, VH FR1, VH CDR1, VH FR2, VH CDR2, VH FR3, VH CDR3, VH FR4, EC, VL, VL FR1, VL CDR1, VL FR2, VL CDR2, VL FR3, VL CDR3, and VL FR4 for individual antibodies, and the SEQ ID NOs assigned to the nucleic acid sequences of the VH and VL of individual antibodies.
Variations of the Disclosed Antibodies and Polynucleotide Sequences Encoding Such Variations [0400] In one embodiment, disclosed herein are anti-CoV-S antibodies or antigen-binding antibody fragments comprising (i) a VH CDR that is same as the VH CDR3 of, (ii) a VH CDR3 and VL CDR3, both of which as same as both of the VH CDR3 and the VL CDR3 of, (iii) at least 1, 2, 3, 4, 5, or 6 CDRs that are same as the corresponding CDR(s) of, or (iv) 6 CDRs that are all the same as the 6 CDRs of any one of the disclosed antibodies selected from the group consisting of ADI-58120, ADI- 58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI- 58129, ADI-58130, and ADI-58131.
[0401] In further embodiments, disclosed herein are anti-CoV-S antibodies or antigen-binding antibody fragments which optionally may be affinity-matured, comprising one of the CDR requirements (i)-(iv) of the immediately above paragraph, further wherein (a) the VH comprises an amino acid sequence with at least 80, 85, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of the VH of, and (b) the VL comprises an amino acid sequence with at least 80, 85, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid
sequence of the VL of any one of the disclosed antibodies selected from the group consisting of ADI- 58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI- 58128, ADI-58129, ADI-58130, and ADI-58131.
[0402] In further embodiments, the disclosure contemplates anti-CoV-S antibodies or antigen binding antibody fragments which optionally may be affinity-matured, comprising one of the VH and VL requirements (i)-(iv) of the immediately above paragraph, further wherein (a) the heavy chain comprises an amino acid sequence with at least 80, 85, 90, 91, 92, 93, 9495, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of the heavy chain of, and (b) the light chain comprises an amino acid sequence with at least 80, 85, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% sequence identity to the amino acid sequence of the light chain of any one of the disclosed antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI- 58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, and ADI-58131.
[0403] In further embodiments, the disclosure contemplates anti-CoV-S antibodies or antigen binding antibody fragments which optionally may be affinity-matured, comprising one of the CDR requirements (i)-(iv) of the immediately above paragraph, further wherein (a) the VH is identical to the VH of, and (b) the VL is identical to the VL of any one of the disclosed antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, and ADI-58131.
[0404] In other embodiments, the disclosure includes antibodies and antigen-binding fragments which optionally may be affinity-matured, having binding specificity to COV-S that bind the same epitope as one of antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI- 58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI- 58130, and ADI-58131.
[0405] In other embodiments, the disclosure includes antibodies and antigen-binding fragments having binding specificity to COV-S, which optionally may be affinity-matured, that bind the same epitope as any one of antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI- 58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI- 58130, and ADI-58131.
[0406] In other embodiments, the anti-CoV-S antibodies and antigen-binding fragments optionally may be affinity-matured, comprise, or alternatively consist of, combinations of one or more of the FRs, CDRs, the VH and VL sequences, and the heavy chain and light chain sequences set forth above, including all of them, or sequences that are at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical thereto.
[0407] In a further embodiment, antigen-binding fragments comprise, or alternatively consist of, Fab fragments having binding specificity for CoV-S. The Fab fragment preferably includes the VH and the VL sequence of any one of antibodies selected from the group consisting of ADI-58120, ADI-
58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI- 58129, ADI-58130, and ADI-58131, or sequences that are at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical thereto. This embodiment further includes Fabs containing additions, deletions, and variants of such VH and VL sequence while retaining binding specificity for CoV-S.
[0408] In some embodiments, Fab fragments may be produced by enzymatic digestion (e.g., papain) of the parent full antibody. In another embodiment, anti-CoV-S antibodies, such as anyone of antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, and ADI- 58131, and Fab fragments thereof may be produced via expression in mammalian cells, such as CHO, NSO, or HEK 293 cells, fungal, insect, or microbial systems, such as yeast cells.
[0409] In additional embodiments, disclosed herein are polynucleotides encoding antibody polypeptides having binding specificity to COV-S, including the VH and VL of any one of antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, and ADI-58131, as well as fragments, variants, optionally affinity-matured variants, and combinations of one or more of the FRs, CDRs, the VH and VL sequences, and the heavy chain and light chain sequences set forth above, including all of them, or sequences that are at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%,
98%, or 99% identical thereto.
[0410] In other embodiments, the disclosure contemplates isolated anti-CoV-S antibodies and antigen binding fragments comprising (i) a VH which is same as the VH of any one of antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, and ADI-58131; and (ii) a VL which is same as the VL of another antibody selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, and ADI-58131, or a variant thereof, wherein optionally one or more of the framework region residues (“LR residues”) and/or CDR residues in said VH or VL polypeptide has been substituted with another amino acid residue resulting in an anti-CoV -S antibody that specifically binds COV-S.
[0411] The disclosure also includes humanized, primatized and other chimeric forms of these antibodies. The chimeric and humanized antibodies may include an Lc derived from IgGl, IgG2,
IgG3, or IgG4 constant regions.
[0412] In some embodiments, the chimeric or humanized antibodies or fragments or VH or VL polypeptides originate or are derived from one or more human antibodies, e.g., a human antibody identified from a clonal human B cell population.
[0413] In some aspects, the disclosure provides vectors comprising a nucleic acid molecule encoding an anti-CoV-S antibody or fragment thereof as disclosed herein. In some embodiments, the disclosure provides host cells comprising a nucleic acid molecule encoding an anti-CoV-S antibody or fragment thereof as disclosed herein.
[0414] In some aspects, the disclosure provides isolated antibodies or antigen binding fragments thereof that competes for binding to CoV-S with an antibody or antigen binding fragment thereof disclosed herein.
[0415] In some aspects, the disclosure provides a nucleic acid molecule encoding any of the antibodies or antigen binding fragments disclosed herein.
[0416] In some aspects, the disclosure provides a pharmaceutical or diagnostic composition comprising at least one antibody or antigen binding fragment thereof as disclosed herein.
[0417] In some aspects, the disclosure provides a method for treating or preventing a condition associated with elevated CoV-S levels in a subject, comprising administering to a subject in need thereof an effective amount of at least one isolated antibody or antigen binding fragment thereof as disclosed herein.
[0418] In some aspects, the disclosure provides a method of inhibiting binding of COV-S to its receptor (e.g., ACE2, L-SIGN, CD209L, DPP4, CD26) or an S protein-priming protein (e.g., TMPRSS2) in a subject comprising administering an effective amount of at least one antibody or antigen binding fragment thereof as disclosed herein. For example, administering one or more of antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, and ADI- 58131 may inhibit binding of COV-S to its receptor, e.g., ACE2.
[0419] In some aspects, the disclosure provides an antibody or antigen binding fragment thereof that selectively binds to CoV-S, wherein the antibody or antigen binding fragment thereof binds to CoV-S with a KD of less than or equal to 5x105 M, 105 M, 5x106 M, 106 M, 5x107 M, 107 M, 5x108 M,
108 M, 5x109 M, 109 M, 5x1010 M, 1010 M, 5x1011 M, 1011 M, 5x1012 M, 1012 M, 5x1013 M, or 1013 M; preferably, with a KD of less than or equal to 5x1010 M, 1010 M, 5x1011 M, 1011 M, 5x1012 M, or 1012 M; more preferably, with a KD that is less than about 100 pM, less than about 50 pM, less than about 40 pM, less than about 25 pM, less than about 1 pM, between about 10 pM and about 100 pM, between about 1 pM and about 100 pM, or between about 1 pM and about 10 pM. Preferably, the anti-CoV-S antibody or antigen binding fragment has cross-reactivity to the S protein of CoV other than SARS-CoV-S or SARS-CoV-2-S.
[0420] The inventive antibodies and antigen binding fragments thereof may be modified post- translationally to add effector moieties such as chemical linkers, detectable moieties such as for example fluorescent dyes, enzymes, substrates, bioluminescent materials, radioactive materials, and
chemiluminescent moieties, or functional moieties such as for example streptavidin, avidin, biotin, a cytotoxin, a cytotoxic agent, and radioactive materials.
[0421] Antibodies and antigen binding fragments thereof may also be chemically modified to provide additional advantages such as increased solubility, stability and circulating time (in vivo half- life) of the polypeptide, or decreased immunogenicity ( See U.S. Patent No. 4,179,337). The chemical moieties for derivatization may be selected from water soluble polymers such as polyethylene glycol, ethylene glycol/propylene glycol copolymers, carboxymethylcellulose, dextran, polyvinyl alcohol, and the like. The antibodies and fragments thereof may be modified at random positions within the molecule, or at predetermined positions within the molecule and may include one, two, three, or more attached chemical moieties.
[0422] The polymer may be of any molecular weight, and may be branched or unbranched. For polyethylene glycol, the preferred molecular weight is between about 1 kDa and about 100 kDa (the term "about" indicating that in preparations of polyethylene glycol, some molecules will weigh more, some less, than the stated molecular weight) for ease in handling and manufacturing. Other sizes may be used, depending on the desired therapeutic profile (e.g., the duration of sustained release desired, the effects, if any on biological activity, the ease in handling, the degree or lack of antigenicity and other known effects of the polyethylene glycol to a therapeutic protein or analog). For example, the polyethylene glycol may have an average molecular weight of about 200, 500, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 5500, 6000, 6500, 7000, 7500, 8000, 8500, 9000, 9500, 10,000,
10.500, 11,000, 11,500, 12,000, 12,500, 13,000, 13,500, 14,000, 14,500, 15,000, 15,500, 16,000,
16.500, 17,000, 17,500, 18,000, 18,500, 19,000, 19,500, 20,000, 25,000, 30,000, 35,000, 40,000,
50,000, 55,000, 60,000, 65,000, 70,000, 75,000, 80,000, 85,000, 90,000, 95,000, or 100,000 kDa.
Branched polyethylene glycols are described, for example, in U.S. Patent No. 5,643,575; Morpurgo et al., Appl. Biochem. Biotechnol., 56:59-72 (1996); Vorobjev et al, Nucleosides and Nucleotides, 18:2745-2750 (1999); and Caliceti et al, Bioconjug. Chem., 10:638-646 (1999), the disclosures of each of which are incorporated herein by reference.
[0423] There are a number of attachment methods available to those skilled in the art ( See e.g., EP 0 401 384, herein incorporated by reference, disclosing a method of coupling PEG to G-CSF; and Malik et al, Exp. Hematol, 20:1028-1035 (1992) (reporting pegylation of GM-CSF using tresyl chloride)). For example, polyethylene glycol may be covalently bound through amino acid residues via a reactive group, such as, a free amino or carboxyl group. Reactive groups are those to which an activated polyethylene glycol molecule may be bound. The amino acid residues having a free amino group may include lysine residues and the N-terminal amino acid residues; those having a free carboxyl group may include aspartic acid residues glutamic acid residues and the C-terminal amino acid residue. Sulfhydryl groups may also be used as a reactive group for attaching the polyethylene glycol
molecules. Preferred for therapeutic purposes is attachment at an amino group, such as attachment at the N-terminus or lysine group.
[0424] As described above, polyethylene glycol may be attached to proteins via linkage to any of a number of amino acid residues. For example, polyethylene glycol can be linked to polypeptides via covalent bonds to lysine, histidine, aspartic acid, glutamic acid, or cysteine residues. One or more reaction chemistries may be employed to attach polyethylene glycol to specific amino acid residues (e.g., lysine, histidine, aspartic acid, glutamic acid, or cysteine) or to more than one type of amino acid residue (e.g., lysine, histidine, aspartic acid, glutamic acid, cysteine and combinations thereof). [0425] Alternatively, antibodies or antigen binding fragments thereof having increased in vivo half- lives may be produced via fusion with albumin (including but not limited to recombinant human serum albumin or fragments or variants thereof (See, e.g., U.S. Patent No. 5,876,969, EP 0413 622, and U.S. Patent No. 5,766,883, herein incorporated by reference in their entirety)), or other circulating blood proteins such as transferrin or ferritin. In a preferred embodiment, polypeptides and/or antibodies of the present disclosure (including fragments or variants thereof) are fused with the mature form of human serum albumin (i.e., amino acids 1-585 of human serum albumin as shown in FIGS. 1 and 2 of EP 0 322094) which is herein incorporated by reference in its entirety. Polynucleotides encoding fusion proteins of the disclosure are also encompassed by the disclosure. [0426] Regarding detectable moieties, further exemplary enzymes include, but are not limited to, horseradish peroxidase, acetylcholinesterase, alkaline phosphatase, Z¾Za-galactosidase, and luciferase. Further exemplary fluorescent materials include, but are not limited to, rhodamine, fluorescein, fluorescein isothiocyanate, umbelliferone, dichlorotriazinylamine, phycoerythrin, and dansyl chloride. Further exemplary chemiluminescent moieties include, but are not limited to, luminol. Further exemplary bioluminescent materials include, but are not limited to, luciferin and aequorin. Further exemplary radioactive materials include, but are not limited to, Iodine 125 (125I), Carbon 14 (14C), Sulfur 35 (35S), Tritium (3H) and Phosphorus 32 (32P).
[0427] Methods are known in the art for conjugating an antibody or antigen binding fragment thereof to a detectable moiety and the like, such as for example those methods described by Hunter et al, Nature, 144:945 (1962); David et al, Biochemistry, 13:1014 (1974); Pain et al, J. Immunol. Meth., 40:219 (1981); and Nygren, J., Histochem. and Cytochem., 30:407 (1982).
[0428] Embodiments described herein further include variants and equivalents that are substantially homologous to the antibodies, antibody fragments, diabodies, SMIPs, camelbodies, nanobodies, IgNAR, polypeptides, variable regions, and CDRs set forth herein. These may contain, e.g., conservative substitution mutations, (i.e., the substitution of one or more amino acids by similar amino acids). For example, conservative substitution refers to the substitution of an amino acid with another within the same general class, e.g., one acidic amino acid with another acidic amino acid, one
basic amino acid with another basic amino acid, or one neutral amino acid by another neutral amino acid. The intent of a conservative amino acid substitution is well known in the art.
[0429] In other embodiments, the disclosure contemplates polypeptide sequences having at least 90% or greater sequence homology to any one or more of the polypeptide sequences of antigen binding fragments, variable regions and CDRs set forth herein. More preferably, the disclosure contemplates polypeptide sequences having at least 95% or greater sequence homology, even more preferably at least 98% or greater sequence homology, and still more preferably at least 99% or greater sequence homology to any one or more of the polypeptide sequences of antigen binding fragments, variable regions, and CDRs set forth herein.
[0430] Methods for determining homology between nucleic acid and amino acid sequences are well known to those of ordinary skill in the art.
[0431] In other embodiments, the disclosure further contemplates the above-recited polypeptide homologs of the antigen binding fragments, variable regions and CDRs set forth herein further having anti-CoV-S activity. Non-limiting examples of anti-CoV-S activity are set forth herein, e.g., ability to inhibit CoV-S binding to its receptor such as ACE2 or L-SIGN or an S protein-priming protein, thereby resulting in the reduced entry of CoV into cells.
[0432] In other embodiments, the disclosure further contemplates the generation and use of antibodies that bind any of the foregoing sequences, including, but not limited to, anti-idiotypic antibodies. In an exemplary embodiment, such an anti-idiotypic antibody could be administered to a subject who has received an anti-CoV-S antibody to modulate, reduce, or neutralize, the effect of the anti-CoV-S antibody. Such antibodies could also be useful for treatment of an autoimmune disease characterized by the presence of anti-CoV-S antibodies. A further exemplary use of such antibodies, e.g., anti-idiotypic antibodies, is for detection of the anti-CoV-S antibodies of the present disclosure, for example to monitor the levels of the anti-CoV-S antibodies present in a subject’s blood or other bodily fluids. For example, in one embodiment, the disclosure provides a method of using the anti- idiotypic antibody to monitor the in vivo levels of said anti-CoV-S antibody or antigen binding fragment thereof in a subject or to neutralize said anti-CoV-S antibody in a subject being administered said anti-CoV-S antibody or antigen binding fragment thereof.
[0433] The present disclosure also contemplates anti-CoV-S antibodies comprising any of the polypeptide or polynucleotide sequences described herein substituted for any of the other polynucleotide sequences described herein. For example, without limitation thereto, the present disclosure contemplates antibodies comprising the combination of any of the VF and VH sequences described herein, and further contemplates antibodies resulting from substitution of any of the CDR sequences described herein for any of the other CDR sequences described herein.
[0434] Another embodiment of the disclosure contemplates these polynucleotides incorporated into an expression vector for expression in mammalian cells such as CHO, NSO, or HEK-293 cells, or in
fungal, insect, or microbial systems such as yeast cells. In one embodiment of the disclosure described herein, Fab fragments can be produced by enzymatic digestion (e.g., papain) of any one of antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, and ADI-58131, following expression of the full-length polynucleotides in a suitable host. In another embodiment, anti-Co V-S antibodies, such as anyone of antibodies selected from the group consisting of ADI- 58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI- 58128, ADI-58129, ADI-58130, and ADI-58131, or Fab fragments thereof, can be produced via expression of the polynucleotides encoding any one of antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI- 58127, ADI-58128, ADI-58129, ADI-58130, and ADI-58131, in mammalian cells such as CFIO,
NSO, or F1EK 293 cells, fungal, insect, or microbial systems such as yeast cells.
[0435] Flost cells and vectors comprising said polynucleotides are also contemplated.
[0436] The disclosure further contemplates vectors comprising the polynucleotide sequences encoding the variable heavy and light chain polypeptide sequences, as well as the individual CDRs (hypervariable regions), as set forth herein, as well as host cells comprising said vector sequences. In one embodiment, the host cells are mammalian cells, such as CFIO cells. In one embodiment, the host cells are yeast cells.
D. Antibody -drug Conjugate Comprising Anti-CoV-S Antibody
[0437] In some aspects, the disclosure is further directed to antibody-drug conjugates (ADCs) comprising (a) any antibody or antigen-binding antibody fragment described herein; and (b) a drug conjugated to the antibody or antigen-binding antibody fragment, either directly or indirectly (e.g., via a linker).
[0438] In some aspects, the drug may be, but not limited to, a cytotoxic drug, an apoptotic drug, an immunostimulatory drug, an anti-microbial drug, an antibacterial drug or vaccine, an antiviral drug, antihelminth drug, antiparasitic drug, an anti-inflammatory drug, antihistamine, an anti-fibrotic drug, an immunosuppressive drug, a steroid, a bronchodilator, a beta blocker, an ACE inhibitor, an enzyme, a serine protease inhibitor, a toxin, a radioisotope, a compound, a small molecule, a small molecule inhibitor, a protein, a peptide, a vector, a plasmid, a viral particle, a nanoparticle, a DNA molecule, an RNA molecule, an siRNA, an shRNA, a micro RNA, an oligonucleotide, and an imaging drug.
[0439] An antiviral drug may be remdesivir, favipiravir, darunavir, nelfinavir, saquinavir, lopinavir or ritonavir; an antihelminth drug may be ivermectin; an antiparasite drug may be hydroxychloroquine, chloroquine, or atovaquone; antibacterial drug or vaccine may be the tuberculosis vaccine BCG; an anti-inflammatory drug, may be ciclesonide, a TNF inhibitor (e.g., adalimumab), a TNF receptor inhibitor (e.g., etanercept), an IL-6 inhibitor (e.g., clazakizumab), an
IL-6 receptor inhibitor (e.g., toclizumab), or metamizole; an antihistamine drug may be bepotastine; an ACE inhibitor may be moexipril; and a drug that inhibits priming of CoV-S may be a serine protease inhibitor such as nafamostat.
[0440] The toxin may be a bacterial, fungal, plant, or animal toxin, or a fragment thereof. Examples include, but are not limited to, diphtheria A chain, diphtheria toxin, exotoxin A chain, ricin A chain, abrin A chain, modeccin A chain, alpha sarcin, Aleurites fordii protein, a dianthin protein, or a Phytolacca Americana protein.
[0441] The cytotoxic drug or anti-proliferative drug may be, for example, but is not limited to, doxorubicin, daunorubicin, cucurbitacin, chaetocin, chaetoglobosin, chlamydocin, calicheamicin, nemorubicin, cryptophyscin, mensacarcin, ansamitocin, mitomycin C, geldanamycin, mechercharmycin, rebeccamycin, safracin, okilactomycin, oligomycin, actinomycin, sandramycin, hypothemycin, polyketomycin, hydroxyellipticine, thiocolchicine, methotrexate, triptolide, taltobulin, lactacystin, dolastatin, auristatin, monomethyl auristatin E (MMAE), monomethyl auristatin F (MMAF), telomestatin, tubastatin A, combretastatin, maytansinoid, MMAD, MMAF, DM1, DM4, DTT, 16-GMB -APA-GA, 17-DMAP-GA, JW 55, pyrrolobenzodiazepine, SN-38, Ro 5-3335, puwainaphycin, duocarmycin, bafilomycin, taxoid, tubulysin, ferulenol, lusiol A, fumagillin, hygrolidin, glucopiericidin, amanitin, ansatrienin, cinerubin, phallacidin, phalloidin, phytosphongosine, piericidin, poronetin, phodophyllotoxin, gramicidin A, sanguinarine, sinefungin, herboxidiene, microcolin B, microcystin, muscotoxin A, tolytoxin, tripolin A, myoseverin, mytoxin B, nocuolin A, psuedolaric acid B, pseurotin A, cyclopamine, curvulin, colchicine, aphidicolin, englerin, cordycepin, apoptolidin, epothilone A, limaquinone, isatropolone, isofistularin, quinaldopeptin, ixabepilone, aeroplysinin, arruginosin, agrochelin, epothilone, or a derivative thereof (for example, see Polakis P. et ai, Pharmacol Rev. 2016 Jan;68(l):3-19. doi: 10.1124/pr.114.009373) (the drugs may be obtained from many vendors, including Creative Biolabs ®).
[0442] The radioisotope may be for example, but is not limited to, At211, 1131, In131, 1125, Y90, Re186, Re188, Sm153, Bi212, P32, Pb212 and radioactive isotopes of Lu.
[0443] In certain embodiments, the drug may be, but is not limited to, MMAE or MMAF.
[0444] In some embodiments, the Ab or antigen-binding Ab fragment is directly conjugated to the drug to form an ADC.
[0445] In some embodiments, the antibody or antigen-binding antibody fragment is indirectly conjugated to the drug to form an ADC.
[0446] Any appropriate conjugation method may be used to generate an ADC (for example, Nolting B. Methods Mol Biol. 2013;1045:71-100; Jain N. etal., Pharm Res. 2015 Nov;32(ll):3526-40; Tsuchikama K. et al, Protein Cell. 2018 Jan;9(l):33-46; Polakis P. et ai, Pharmacol Rev . 2016 Jan;68(l):3-19). Examples of methods that may be used to perform conjugation include, but are not limited to, chemical conjugation and enzymatic conjugation.
[0447] Chemical conjugation may utilize, for example, but is not limited to, lysine amide coupling, cysteine coupling, and/or non-natural amino acid incorporation by genetic engineering. Enzymatic conjugation may utilize, for example, but is not limited to, transpeptidation using sortase, transpeptidation using microbial transglutaminase, and/or N-Glycan engineering.
[0448] In certain aspects, one or more of cleavable linkers may be used for conjugation. The cleavable linker may enable cleavage of the drug upon responding to, for example, but not limited to, an environmental difference between the extracellular and intracellular environments (pH, redox potential, etc.) or by specific lysosomal enzymes.
[0449] Examples of the cleavable linker include, but are not limited to, hydrazone linkers, peptide linkers including cathepsin B-responsive linkers, such as valine -citrulline (vc) linker, disulfide linkers such as N-succinimidyl-4-(2-pyridyldithio) (SPP) linker or N-succinimidyl-4-(2- pyridyldithiojbutanoate (SPDB) linker, and pyrophosphate diester linkers.
[0450] Alternatively or simultaneously, one or more of non-cleavable linkers may be used. Examples of non-cleavable linkers include thioether linkers, such as N-succinimidyl 4-(N-maleimidomethyl) cyclohexane- 1-carboxylate (SMCC), and maleimidocaproyl (me) linkers. Generally, non-cleavable linkers are more resistant to proteolytic degradation and more stable compared to cleavable linkers.
E. Chimeric Antisen Receptor Comprising Anti-CoV-S Antigen-Binding Antibody Frasment [0451] In some embodiments, a compound specific to CoV-S according to the present disclosure may be a chimeric antigen receptor (CAR). In particular, the CARs of the present disclosure comprise an antigen binding (AB) domain that binds to CoV-S, a transmembrane (TM) domain, and an intracellular signaling (ICS) domain. In some embodiments, a CAR may comprise a hinge that joins the AB domain and said TM domain. In some embodiments, the CAR may comprise one or more costimulatory (CS) domains.
AB domain
[0452] A CAR according to the disclosure will comprise an antigen-binding (AB) domain which binds to COV-S. In some embodiments, the AB domain of the CAR may comprise any of the anti- COV-S antigen-binding antibody fragments disclosed herein.
[0453] In some embodiments, the AB domain of the CAR may comprise any of the antigen-binding domain of any of the anti-COV-S antibodies disclosed herein.
[0454] In some embodiments, the AB domain of the CAR may comprise any of the anti-COV-S antibodies, anti-COV-S antigen-binding antibody fragments, anti-COV-S multi-specific Abs, anti- COV-S multi-specific antigen-binding antibody fragments, and anti-COV-S ADCs disclosed herein, or the ABD thereof.
[0455] In some embodiments, the AB domain of the CAR may comprise an anti-COV-S scFv.
[0456] In some embodiments, the AB domain may comprise an amino acid sequence at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical to an scFv comprising the VH and VL of any one of antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI- 58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI- 58130, and ADI-58131.
[0457] In some aspects, the AB domain may compete for binding to COV-S with any one of antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, and ADI- 58131.
Hinge
[0458] In some embodiments, the CAR may comprise a hinge sequence between the AB domain and the TM domain. One of the ordinary skill in the art will appreciate that a hinge sequence is a short sequence of amino acids that facilitates flexibility (see, e.g. Woof J.M. et al, Nat. Rev. Immunol.,
4(2): 89-99 (2004)). The hinge sequence can be any suitable sequence derived or obtained from any suitable molecule.
[0459] In some embodiments, the length of the hinge sequence may be optimized based on the desired length of the extracellular portion of the CAR, which may be based on the location of the epitope within the target molecule. For example, if the epitope is in the membrane proximal region within the target molecule, longer hinges may be optimal.
[0460] In some embodiments, the hinge may be derived from or include at least a portion of an immunoglobulin Fc region, for example, an IgGl Fc region, an IgG2 Fc region, an IgG3 Fc region, an IgG4 Fc region, an IgE Fc region, an IgM Fc region, or an IgA Fc region. In certain embodiments, the hinge includes at least a portion of an IgGl, an IgG2, an IgG3, an IgG4, an IgE, an IgM, or an IgA immunoglobulin Fc region that falls within its CH2 and CH3 domains. In some embodiments, the hinge may also include at least a portion of a corresponding immunoglobulin hinge region. In some embodiments, the hinge is derived from or includes at least a portion of a modified immunoglobulin Fc region, for example, a modified IgGl Fc region, a modified IgG2 Fc region, a modified IgG3 Fc region, a modified IgG4 Fc region, a modified IgE Fc region, a modified IgM Fc region, or a modified IgA Fc region. The modified immunoglobulin Fc region may have one or more mutations (e.g., point mutations, insertions, deletions, duplications) resulting in one or more amino acid substitutions, modifications, or deletions that cause impaired binding of the hinge to an Fc receptor (FcR). In some aspects, the modified immunoglobulin Fc region may be designed with one or more mutations which result in one or more amino acid substitutions, modifications, or deletions that cause impaired binding of the hinge to one or more FcR including, but not limited to, FcyRI, FcyR2A, FcyR2Bl, Fcy2B2, Fey 3A, Fey 3B, FceRI, FcsR2, FcaRI, Fca/pR, or FcRn.
[0461] In some aspects, a portion of the immunoglobulin constant region may serve as a hinge between the AB domain, for example scFv or nanobody, and the TM domain. The hinge can be of a length that provides for increased responsiveness of the CAR-expressing cell following antigen binding, as compared to in the absence of the hinge. In some examples, the hinge is at or about 12 amino acids in length or is no more than 12 amino acids in length. Exemplary hinges include those having at least about 10 to 229 amino acids, about 10 to 200 amino acids, about 10 to 175 amino acids, about 10 to 150 amino acids, about 10 to 125 amino acids, about 10 to 100 amino acids, about 10 to 75 amino acids, about 10 to 50 amino acids, about 10 to 40 amino acids, about 10 to 30 amino acids, about 10 to 20 amino acids, or about 10 to 15 amino acids, and including any integer between the endpoints of any of the listed ranges. In some embodiments, a hinge has about 12 amino acids or less, about 119 amino acids or less, or about 229 amino acids or less. Exemplary hinges include a CD28 hinge, IgG4 hinge alone, IgG4 hinge linked to CH2 and CH3 domains, or IgG4 hinge linked to the CH3 domain. Exemplary hinges include, but are not limited to, those described in Hudecek M. et al. (2013) Clin. Cancer Res., 19:3153, international patent application publication number WO2014031687, U.S. Pat. No. 8,822,647 or published App. No. US2014/0271635.
[0462] Known hinge sequences include those derived from CD8 a molecule or a CD28 molecule.
Transmembrane (TM) domain
[0463] With respect to the TM domain, the CAR can be designed to comprise a TM domain that is fused to the AB domain of the CAR. A hinge sequence may be inserted between the AB domain and the TM domain. TM domains may be derived from a natural or from synthetic sources. Where the source is natural, the domain may be derived from any membrane-bound or transmembrane protein. Typically, a TM domain denotes a single transmembrane a helix of a transmembrane protein, also known as an integral protein. TM domains e.g., may be derived from (i.e. comprise at least the transmembrane region(s) of) CD28, CD3 e, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD45, CD64, CD80, CD86, CD134, CD137, CD154, TCR a, TCR b, or CD3 zeta and/or TM domains containing functional variants thereof such as those retaining a substantial portion of the structural, e.g., transmembrane, properties thereof.
[0464] Alternatively, the TM domain may be synthetic, in which case the TM domain will comprise predominantly hydrophobic residues such as leucine and valine. Preferably a triplet of phenylalanine, tryptophan and valine will be found at each end of a synthetic TM domain. A TM domain is generally thermodynamically stable in a membrane. It may be a single a helix, a transmembrane b barrel, a b- helix of gramicidin A, or any other structure. Transmembrane helices are usually about 20 amino acids in length.
[0465] A well-used TM domain comprises the TM region of CD28, e.g., human CD28. Often, a short oligo- or polypeptide spacer, e.g., between 2 and 10 amino acids in length is used to form the linkage between the TM domain and the ICS domain(s) of the CAR.
Intracellular signaling ) domain and costimulatorv (CS ) domain [0466] The ICS domain or the cytoplasmic domain of a CAR generally triggers or elicits activation of at least one of the normal effector functions of the cell in which the CAR has been placed. The term "effector function" refers to a specialized function of a cell. Effector function of a T cell, for example, may be cytolytic activity or helper activity including the secretion of cytokines. Thus, the term "intracellular signaling domain" or “ICS domain” refers to the portion of a protein which transduces the effector function signal and directs the cell to perform a specialized function. While usually the entire ICS domain can be employed, in many cases it is not necessary to use the entire chain. To the extent that a truncated portion of the intracellular signaling domain is used, such truncated portion may be used in place of the intact chain as long as it transduces the effector function signal. The term "intracellular signaling domain" or “ICS domain” is thus meant to include any truncated portion of the ICS domain sufficient to transduce the effector function signal.
[0467] Examples of known ICS domains include the cytoplasmic sequences of the T cell receptor (TCR) and co-receptors that act in concert to initiate signal transduction following antigen receptor engagement, as well as any derivative or variant of these sequences and any synthetic sequence that has the same functional capability.
[0468] Signals generated through one ICS domain alone may be insufficient for full activation of a cell, and a secondary or costimulatory signal may also be required. In such cases, a costimulatory domain (CS domain) may be included in the cytoplasmic portion of a CAR. A CS domain is a domain that transduces such a secondary or costimulatory signal. In some instances, a CAR of the present disclosure may comprise two or more CS domains. The CS domain(s) may be placed upstream of the ICS domain or downstream of the ICS domain.
[0469] T cell activation can be mediated by two distinct classes of cytoplasmic signaling sequence: those that initiate antigen-dependent primary activation through the TCR (primary cytoplasmic signaling sequences) and those that act in an antigen-independent manner to provide a secondary or costimulatory signal (secondary cytoplasmic signaling sequences). Primary cytoplasmic signaling sequences regulate primary activation of the TCR complex either in a stimulatory way, or in an inhibitory way. Primary cytoplasmic signaling sequences that act in a stimulatory manner may contain signaling motifs which are known as immunoreceptor tyrosine-based activation motifs or IT AMs. Such a cytoplasmic signaling sequence may be contained in the ICS or the CS domain of a CAR. [0470] Examples of ITAM-containing primary cytoplasmic signaling sequences include those derived from an ICS domain of a lymphocyte receptor chain, a TCR/CD3 complex protein, an Fc receptor subunit, an IL-2 receptor subunit, CD3z, FcR g, FcR , CD3y, CD35, CD3e, CD5, CD22, CD66d, CD79a, CD79b, CD278 (ICOS), Fee RI, DAP10, and DAP12. A well-used ICS domain comprises a cytoplasmic signaling sequence derived from CD3 zeta. In some instances, the CD3z ICS domain may be combined with one or more of other cytoplasmic domain(s). For example, the
cytoplasmic domain of the CAR can comprise a CD3 z ICS domain and a CS domain wherein a CS region refers to a portion of the CAR comprising the intracellular domain of a costimulatory molecule. A costimulatory molecule is a cell surface molecule other than an antigen receptor or their ligands that is required for an efficient response of lymphocytes to an antigen.
[0471] Examples of co-stimulatory molecules include an MHC class I molecule, TNF receptor proteins, Immunoglobulin-like proteins, cytokine receptors, integrins, signaling lymphocytic activation molecules (SLAM proteins), activating NK cell receptors, a Toll ligand receptor, B7-H3, BAFFR, BTLA, BLAME (SLAMF8), CD2, CD4, CD5, CD7, CD 8 a, CD 8 b, CDlla, LFA-1 (CDlla/CD18), CDllb, CDllc, CDlld, CD18, CD19, CD19a, CD27, CD28, CD29, CD30, CD40, CD49a, CD49D, CD49f, CD69, CD84, CD96 (Tactile), CD100 (SEMA4D), CD103, CRTAM, 0X40 (CD 134), 4-1BB (CD137), SLAM (SLAMF1, CD150, IPO-3), CD160 (BY55), SELPLG (CD162), DNAM1 (CD226), Ly9 (CD229), SLAMF4 (CD244, 2B4), ICOS (CD278), CEACAM1, CDS, CRTAM, DAP 10, GADS, GITR, HVEM (LIGHTR), IA4, ICAM-1, IL2R b, IL2R g, IL7R a, ITGA4, ITGA6, ITGAD, ITGAE, ITGAL, ITGAM, ITGAX, ITGB1, ITGB2, ITGB7, KIRDS2, LAT, LFA-1, LIGHT, LTBR, NKG2C, NKG2D, NKp30, NKp44, NKp46, NKp80 (KLRF1), PAG/Cbp, PD-1, PSGL1, SLAMF6 (NTB-A, Lyl08), SLAMF7, SLP-76, TNFR2, TRANCE/RANKL, VLA1, VLA-6, a ligand that specifically binds with CD83, and the like. The ICS domain and the CS domain(s) of the CAR may be linked to each other in a random or specified order, optionally via a short oligo- or polypeptide linker, e.g., between 2 and 10 amino acids in length.
Exemplary CAR constructs
[0472] A CAR construct may comprise the following format: “AB domain - hinge - TM domain - CS domain - ICS domain.”
[0473] CARs may comprise an amino acid sequence at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical to any of the exemplary constructs below. In the exemplary constructs below, the “anti-CoV-S scFv” may be an scFv generated by linking the VH and VL (in the order of VH-linker-VL or VL-linker-VH) of any one of anti-CoV-S antibodies selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, and ADI-58131.
[0474] In some embodiments, a leader sequence (LS) may be placed upstream of the polynucleotide sequences encoding the CAR. The leader sequence facilitates the expression of the CAR on the cell surface.
Further modification
[0475] CARs according to the present disclosure, nucleotide sequences encoding the same, vectors encoding the same, and cells comprising nucleotide sequences encoding said CARs may be further modified, engineered, optimized, or appended in order to provide or select for various features. These
features may include, but are not limited to, efficacy, persistence, target specificity, reduced immunogenicity, multi-targeting, enhanced immune response, expansion, growth, reduced off-target effect, reduced subject toxicity, improved target cytotoxicity, improved attraction of disease alleviating immune cells, detection, selection, targeting, and the like. For example, the cells may be engineered to express another CAR, or to have a suicide mechanism, and may be modified to remove or modify expression of an endogenous receptor or molecule such as a TCR and/or MHC molecule. [0476] In some embodiments, the vector or nucleic acid sequence encoding the CAR further encodes other genes. The vector or nucleic acid sequence may be constructed to allow for the co-expression of multiple genes using a multitude of techniques including co-transfection of two or more plasmids, the use of multiple or bidirectional promoters, or the creation of bicistronic or multicistronic vectors. The construction of multicistronic vectors may include the encoding of IRES elements or 2 A peptides, such as T2A, P2A, E2A, or F2A (for example, see Kim, J.H., et ai, “High cleavage efficiency of a 2A peptide derived from porcine teschovirus-1 in human cell lines, zebrafish and mice”, PLoS One.
2011 ;6(4)). The CAR expressing cell may further comprise a disruption to one or more endogenous genes.
Efficacy
[0477] The CARs of the present disclosure and cells expressing these CARs may be further modified to improve efficacy against cells expressing the target molecule. The cells may be cells expressing COV-S. The cells expressing COV-S may be cancer cells, vascular cells, or any other target disease- associated cells. In some embodiments, the improved efficacy may be measured by increased cytotoxicity against cells expressing the target molecule, for example cytotoxicity against cancer cells. In some embodiments, the improved efficacy may also be measured by increased production of cytotoxic mediators such as, but not limited to, IFN g, perforin, and granzyme B. In some embodiments, the improved efficacy may be shown by reduction in the signature cytokines of the diseases, or alleviated symptoms of the disease when the CAR expressing cells are administered to a subject. Other cytokines that may be reduced include TGF-beta, IL-6, IL-4, IL-10, and/or IL-13. the improved efficacy may be shown by COV-S-specific immune cell responses, such as T cell cytotoxicity. In case of cancer, improved efficacy may be shown by better tumor cytotoxicity, better infiltration into the tumor, reduction of immunosuppressive mediators, reduction in weight decrease, reduction in ascites, reduction in tumor burden, and/or increased lifespan. In case of autoimmune diseases, reduced responsiveness of autoreactive cells or decrease in autoreactive T cells, B cells, or Abs may represent improved efficacy. In some embodiments, gene expression profiles may be also investigated to evaluate the efficacy of the CAR.
[0478] In one aspect, the CAR expressing cells are further modified to evade or neutralize the activity of immunosuppressive mediators, including, but not limited to prostaglandin E2 (PGE2) and adenosine. In some embodiments, this evasion or neutralization is direct. In other embodiments, this
evasion or neutralization is mediated via the inhibition of protein kinase A (PKA) with one or more binding partners, for example ezrin. In a specific embodiment, the CAR-expressing cells further express the peptide “regulatory subunit I anchoring disruptor” (RIAD). RIAD is thought to inhibit the association of protein kinase A (PKA) with ezrin, which thus prevents PKA’s inhibition of TCR activation (Newick K. et al. Cancer Immunol Res. 2016 Jun;4(6):541-51).
[0479] In some embodiments, the CAR expressing cells may induce a broad immune response, consistent with epitope spreading.
[0480] In some embodiments, the CAR expressing cells further comprise a homing mechanism. For example, the cell may transgenically express one or more stimulatory chemokines or cytokines or receptors thereof. In particular embodiments, the cells are genetically modified to express one or more stimulatory cytokines. In certain embodiments, one or more homing mechanisms are used to assist the inventive cells to accumulate more effectively to the disease site. In some embodiments, the CAR expressing cells are further modified to release inducible cytokines upon CAR activation, e.g., to attract or activate innate immune cells to a targeted cell (so-called fourth generation CARs or TRUCKS). In some embodiments, CARs may co-express homing molecules, e.g., CCR4 or CCR2b, to increase trafficking to the disease site.
Controlling CAR expression
[0481] In some instances, it may be advantageous to regulate the activity of the CAR or CAR expressing cells CAR. For example, inducing apoptosis using, e.g., a caspase fused to a dimerization domain (see, e.g., Di et al, N Engl. J. Med. 2011 Nov. 3; 365(18): 1673-1683), can be used as a safety switch in the CAR therapy of the instant disclosure. In another example, CAR-expressing cells can also express an inducible Caspase-9 (iCaspase-9) molecule that, upon administration of a dimerizer drug (e.g., rimiducid (also called AP1903 (Bellicum Pharmaceuticals) or AP20187 (Ariad)) leads to activation of the Caspase-9 and apoptosis of the cells. The iCaspase-9 molecule contains a chemical inducer of dimerization (CID) binding domain that mediates dimerization in the presence of a CID. This results in inducible and selective depletion of CAR-expressing cells. In some cases, the iCaspase-9 molecule is encoded by a nucleic acid molecule separate from the CAR-encoding vector(s). In some cases, the iCaspase-9 molecule is encoded by the same nucleic acid molecule as the CAR-encoding vector. The iCaspase-9 can provide a safety switch to avoid any toxicity of CAR- expressing cells. See, e.g., Song et al. Cancer Gene Ther. 2008; 15(10):667-75; Clinical Trial Id. No. NCT02107963; and Di et al. N. Engl. J. Med. 2011; 365:1673-83.
[0482] Alternative strategies for regulating the CAR therapy include utilizing small molecules or antibodies that deactivate or turn off CAR activity, e.g., by deleting CAR-expressing cells, e.g., by inducing antibody dependent cell-mediated cytotoxicity (ADCC). For example, CAR-expressing cells described herein may also express an antigen that is recognized by molecules capable of inducing cell death, e.g., ADCC or compliment-induced cell death. For example, CAR expressing cells described
herein may also express a receptor capable of being targeted by an antibody or antibody fragment. Examples of such receptors include EpCAM, VEGFR, integrins (e.g., integrins anb3, a4, aI3/4b3, a4b7, a5b1, anb3, an), members of the TNF receptor superfamily (e.g., TRAIL-R1, TRAIL -R2), PDGF Receptor, interferon receptor, folate receptor, GPNMB, ICAM-1, HLA-DR, CEA, CA-125, MUC1, TAG-72, IL-6 receptor, 5T4, GD2, GD3, CD2, CD3, CD4, CD5, CD11, CDlla/LFA-1, CD15, CD18/ITGB2, CD19, CD20, CD22, CD23/lgE Receptor, CD25, CD28, CD30, CD33, CD38, CD40, CD41, CD44, CD51, CD52, CD62L, CD74, CD80, CD125, CD147/basigin, CD152/CTLA-4, CD154/CD40L, CD195/CCR5, CD319/SLAMF7, and EGFR, and truncated versions thereof (e.g., versions preserving one or more extracellular epitopes but lacking one or more regions within the cytoplasmic domain). For example, CAR-expressing cells described herein may also express a truncated epidermal growth factor receptor (EGFR) which lacks signaling capacity but retains the epitope that is recognized by molecules capable of inducing ADCC, e.g., cetuximab (ERBITUX®), such that administration of cetuximab induces ADCC and subsequent depletion of the CAR- expressing cells (see, e.g., WO2011/056894, and Jonnalagadda et al, “ Gene Ther. 2013; 20(8)853- 860).
[0483] In some embodiments, the CAR cell comprises a polynucleotide encoding a suicide polypeptide, such as for example RQR8. See, e.g., WO2013153391A, which is hereby incorporated by reference in its entirety. In CAR cells comprising the polynucleotide, the suicide polypeptide may be expressed at the surface of a CAR cell. The suicide polypeptide may also comprise a signal peptide at the amino terminus. Another strategy includes expressing a highly compact marker/suicide gene that combines target epitopes from both CD32 and CD20 antigens in the CAR-expressing cells described herein, which binds rituximab, resulting in selective depletion of the CAR-expressing cells, e.g., by ADCC (see, e.g., Philip et al, Blood 2014; 124(8)1277-1287). Other methods for depleting CAR-expressing cells include administration of CAMPATH®, a monoclonal anti-CD52 antibody that selectively binds and targets mature lymphocytes, e.g., CAR-expressing cells, for destruction, e.g., by inducing ADCC. In other embodiments, the CAR-expressing cell can be selectively targeted using a CAR ligand, e.g., an anti-idiotypic antibody. In some embodiments, the anti-idiotypic antibody can cause effector cell activity, e.g., ADCC or ADC activities, thereby reducing the number of CAR- expressing cells. In other embodiments, the CAR ligand, e.g., the anti-idiotypic antibody, can be coupled to an agent that induces cell killing, e.g., a toxin, thereby reducing the number of CAR- expressing cells. Alternatively, the CAR molecules themselves can be configured such that the activity can be regulated, e.g., turned on and off, as described below.
[0484] In some embodiments, a regulatable CAR (RCAR) where the CAR activity can be controlled is desirable to optimize the safety and efficacy of a CAR therapy. In some embodiments, a RCAR comprises a set of polypeptides, typically two in the simplest embodiments, in which the components of a standard CAR described herein, e.g., an AB domain and an ICS domain, are partitioned on
separate polypeptides or members. In some embodiments, the set of polypeptides include a dimerization switch that, upon the presence of a dimerization molecule, can couple the polypeptides to one another, e.g., can couple an AB domain to an ICS domain. Additional description and exemplary configurations of such regulatable CARs are provided herein and in International Publication No. WO 2015/090229, hereby incorporated by reference in its entirety.
[0485] In an aspect, an RCAR comprises two polypeptides or members: 1) an intracellular signaling member comprising an ICS domain, e.g., a primary ICS domain described herein, and a first switch domain; 2) an antigen binding member comprising an AB domain, e.g., that specifically binds a target molecule described herein, as described herein and a second switch domain. Optionally, the RCAR comprises a TM domain described herein. In an embodiment, a TM domain can be disposed on the intracellular signaling member, on the antigen binding member, or on both. Unless otherwise indicated, when members or elements of an RCAR are described herein, the order can be as provided, but other orders are included as well. In other words, in an embodiment, the order is as set out in the text, but in other embodiments, the order can be different. E.g., the order of elements on one side of a transmembrane region can be different from the example, e.g., the placement of a switch domain relative to an ICS domain can be different, e.g., reversed.
[0486] In some embodiments, the CAR expressing immune cell may only transiently express a CAR. For example, the cells may be transduced with mRNA comprising a nucleic acid sequence encoding an inventive CAR. In this vein, the present disclosure also includes an RNA construct that can be directly transfected into a cell. A method for generating mRNA for use in transfection involves in vitro transcription (IVT) of a template with specially designed primers, followed by polyA addition, to produce a construct containing 3' and 5' untranslated sequences ("UTRs"), a 5' cap and/or Internal Ribosome Entry Site (IRES), the nucleic acid to be expressed, and a polyA tail, typically 50-2000 bases in length (SEQ ID NO: 405). RNA so produced can efficiently transfect different kinds of cells. In one embodiment, the template includes sequences for the CAR. In an embodiment, an RNA CAR vector is transduced into a cell by electroporation.
Target specificity
[0487] The CAR expressing cells may further comprise one or more CARs, in addition to the first CAR. These additional CARs may or may not be specific for the target molecule of the first CAR. In some embodiments, the one or more additional CARs may act as inhibitory or activating CARs. In some aspects, the CAR of some embodiments is the stimulatory or activating CAR; in other aspects, it is the costimulatory CAR. In some embodiments, the cells further include inhibitory CARs (iCARs, see Fedorov et al, Sci. Transl. Medicine, 2013 Dec;5(215): 215ral72), such as a CAR recognizing an antigen other than the target molecule of the first CAR, whereby an activating signal delivered through the first CAR is diminished or inhibited by binding of the inhibitory CAR to its ligand, e.g., to reduce off-target effects.
[0488] In some embodiments, the AB domain of the CAR is or is part of an immunoconjugate, in which the AB domain is conjugated to one or more heterologous molecule(s), such as, but not limited to, a cytotoxic agent, an imaging agent, a detectable moiety, a multimerization domain, or other heterologous molecule. Cytotoxic agents include, but are not limited to, radioactive isotopes (e.g., At211, 1131, 1125, Y90, Rel86, Rel88, Sml53, Bi212, P32, Pb212 and radioactive isotopes of Lu); chemotherapeutic agents; growth inhibitory agents; enzymes and fragments thereof such as nucleolytic enzymes; antibiotics; toxins such as small molecule toxins or enzymatically active toxins. In some embodiments, the AB domain is conjugated to one or more cytotoxic agents, such as chemotherapeutic agents or drugs, growth inhibitory agents, toxins (e.g., protein toxins, enzymatically active toxins of bacterial, fungal, plant, or animal origin, or fragments thereof), or radioactive isotopes.
[0489] In some embodiments, to enhance persistence, the cells may be further modified to overexpress pro-survival signals, reverse anti-survival signals, overexpress Bcl-xL, overexpress hTERT, lack Fas, or express a TGF-b dominant negative receptor. Persistence may also be facilitated by the administration of cytokines, e.g., IF-2, IF-7, and IF-15.
F. B-cell Screening and Isolation
[0490] In one embodiment, the present disclosure contemplates the preparation and isolation of a clonal population of antigen-specific B-cells that may be used for isolating at least one CoV-S antigen-specific cell, which can be used to produce a monoclonal antibody against CoV-S, which is specific to a desired CoV-S antigen, or a nucleic acid sequence corresponding to such an antibody. Methods of preparing and isolating said clonal population of antigen-specific B-cells are taught, for example, in U.S. Patent Publication No. US2007/0269868 to Carvalho-Jensen et al, the disclosure of which is herein incorporated by reference in its entirety. Methods of preparing and isolating said clonal population of antigen-specific B-cells are also taught herein in the examples. Methods of “enriching” a cell population by size or density are known in the art. See, e.g., U.S. Patent No. 5,627,052. These steps can be used in addition to enriching the cell population by antigen-specificity.
G. Methods of Producing Antibodies and Fragments Thereof
[0491] In another embodiment, the present disclosure contemplates methods for producing anti-CoV- S antibodies and fragments thereof. Methods of producing antibodies are well known to those of ordinary skill in the art. For example, methods of producing chimeric antibodies are now well known in the art (See, for example, U.S. Patent No. 4,816,567 to Cabilly et al.·, Morrison et al, Proc. Natl. Acad. Sci. U.S.A., 81:8651-55 (1984); Neuberger et al., Nature, 314:268-270 (1985); Boulianne, G.F. et al, Nature, 312:643-46 (1984), the disclosures of each of which are herein incorporated by reference in their entireties).
[0492] As mentioned above, methods of producing humanized antibodies are now well known in the art (See, for example, U.S. Patent Nos. 5,530,101, 5,585,089, 5,693,762, and 6,180,370 to Queen et al U.S. Patent Nos. 5,225,539 and 6,548,640 to Winter; U.S. Patent Nos. 6,054,297, 6,407,213 and 6,639,055 to Carter et al U.S. Patent No. 6,632,927 to Adair; Jones, P.T. et al, Nature, 321:522-525 (1986); Reichmann, L. et al., Nature, 332:323-327 (1988); Verhoeyen, M. etal., Science, 239:1534- 36 (1988), the disclosures of each of which are herein incorporated by reference in their entireties). [0493] Antibody polypeptides of the disclosure having CoV-S binding specificity may also be produced by constructing, using conventional techniques well known to those of ordinary skill in the art, an expression vector containing a promoter (optionally as a component of a eukaryotic or prokaryotic operon) and a DNA sequence encoding an antibody heavy chain in which the DNA sequence encoding the CDRs required for antibody specificity is derived from a non-human cell source, e.g., a rabbit or rodent B-cell source, while the DNA sequence encoding the remaining parts of the antibody chain is derived from a human cell source.
[0494] A second expression vector is produced using the same conventional means well known to those of ordinary skill in the art, said expression vector containing a promoter (optionally as a component of a eukaryotic or prokaryotic operon) and a DNA sequence encoding an antibody light chain in which the DNA sequence encoding the CDRs required for antibody specificity is derived from a non-human cell source, e.g., a rabbit or rodent B-cell source, while the DNA sequence encoding the remaining parts of the antibody chain is derived from a human cell source.
[0495] The expression vectors are transfected into a host cell by convention techniques well known to those of ordinary skill in the art to produce a transfected host cell, said transfected host cell cultured by conventional techniques well known to those of ordinary skill in the art to produce said antibody polypeptides.
[0496] The host cell may be co-transfected with the two expression vectors described above, the first expression vector containing DNA encoding a promoter (optionally as a component of a eukaryotic or prokaryotic operon) and a light chain-derived polypeptide and the second vector containing DNA encoding a promoter (optionally as a component of a eukaryotic or prokaryotic operon) and a heavy chain-derived polypeptide. The two vectors contain different selectable markers, but preferably achieve substantially equal expression of the heavy and light chain polypeptides. Alternatively, a single vector may be used, the vector including DNA encoding both the heavy and light chain polypeptides. The coding sequences for the heavy and light chains may comprise cDNA, genomic DNA, or both.
[0497] The host cells used to express the antibody polypeptides may be either a bacterial cell such as E. coli, or a eukaryotic cell such as P. pastoris. In one embodiment, a mammalian cell of a well- defined type for this purpose, such as a myeloma cell, a CHO cell line, a NSO cell line, or a HEK293 cell line may be used.
[0498] The general methods by which the vectors may be constructed, transfection methods required to produce the host cell and culturing methods required to produce the antibody polypeptides from said host cells all include conventional techniques. Although preferably the cell line used to produce the antibody is a mammalian cell line, any other suitable cell line, such as a bacterial cell line such as an E. coli- derived bacterial strain, or a yeast cell line, may alternatively be used.
[0499] Similarly, once produced the antibody polypeptides may be purified according to standard procedures in the art, such as for example cross-flow filtration, ammonium sulphate precipitation, affinity column chromatography, hydrophobic interaction chromatography (“HIC”), and the like. [0500] The antibody polypeptides described herein may also be used for the design and synthesis of either peptide or non-peptide mimetics that would be useful for the same therapeutic applications as the antibody polypeptides of the disclosure (See, for example, Saragobi et ai, Science, 253:792-795 (1991), the contents of which are herein incorporated by reference in its entirety).
[0501] In another embodiment, the present disclosure contemplates methods for humanizing antibody heavy and light chains which bind to CoV-S. Exemplary methods for humanizing antibody heavy and light chains that may be applied to anti-CoV-S antibodies are identified herein and are conventional in the art.
H. Screening Assays
[0502] The screening assays described here may be used to identify high affinity anti-CoV-S Abs which may be useful in the treatment of diseases and disorders associated with CoV-S in subjects exhibiting symptoms of a CoV-S associated disease or disorder.
[0503] In some embodiments, the antibody is used as a diagnostic tool. The antibody can be used to assay the amount of CoV-S present in a sample and/or subject. As will be appreciated by one of skill in the art, such antibodies need not be neutralizing antibodies. In some embodiments, the diagnostic antibody is not a neutralizing antibody. In some embodiments, the diagnostic antibody binds to a different epitope than the neutralizing antibody binds to. In some embodiments, the two antibodies do not compete with one another.
[0504] In some embodiments, the antibodies disclosed herein are used or provided in an assay kit and/or method for the detection of CoV-S in mammalian tissues or cells in order to screen/diagnose for a disease or disorder associated with changes in levels of CoV-S. The kit comprises an antibody that binds CoV-S and means for indicating the binding of the antibody with CoV-S, if present, and optionally CoV-S protein levels. Various means for indicating the presence of an antibody can be used. For example, fluorophores, other molecular probes, or enzymes can be linked to the antibody and the presence of the antibody can be observed in a variety of ways. The method for screening for such disorders can involve the use of the kit, or simply the use of one of the disclosed antibodies and the determination of whether the antibody binds to CoV-S in a sample. As will be appreciated by one
of skill in the art, high or elevated levels of CoV-S will result in larger amounts of the antibody binding to CoV-S in the sample. Thus, degree of antibody binding can be used to determine how much CoV-S is in a sample. Subjects or samples with an amount of CoV-S that is greater than a predetermined amount (e.g., an amount or range that a person without a CoV-S -related disorder would have) can be characterized as having a CoV-S-mediated disorder.
[0505] The present disclosure further provides for a kit for detecting binding of an anti-CoV-S antibody of the disclosure to CoV-S. In particular, the kit may be used to detect the presence of CoV- S specifically reactive with an anti-CoV-S antibody or an immunoreactive fragment thereof. The kit may also include an antibody bound to a substrate, a secondary antibody reactive with the antigen and a reagent for detecting a reaction of the secondary antibody with the antigen. Such a kit may be an ELISA kit and can comprise the substrate, primary and secondary antibodies when appropriate, and any other necessary reagents such as detectable moieties, enzyme substrates, and color reagents, for example as described herein. The diagnostic kit may also be in the form of an immunoblot kit. The diagnostic kit may also be in the form of a chemiluminescent kit (Meso Scale Discovery,
Gaithersburg, MD). The diagnostic kit may also be a lanthanide -based detection kit (PerkinElmer, San Jose, CA).
[0506] A skilled clinician would understand that a biological sample includes, but is not limited to, sera, plasma, urine, fecal sample, saliva, mucous, pleural fluid, synovial fluid, and spinal fluid.
7. Methods of Ameliorating or Reducing Symptoms of, or Treating, or Preventing, Diseases and Disorders Associated with CoV
[0507] In another embodiment, anti-CoV-S antibodies described herein, or antigen-binding fragments thereof, are useful for ameliorating or reducing the symptoms of, or treating, or preventing, diseases and disorders associated with CoV-S. Anti-CoV-S antibodies described herein, or antigen binding fragments thereof, as well as combinations, can also be administered in a therapeutically effective amount to patients in need of treatment of diseases and disorders associated with CoV-S in the form of a pharmaceutical composition as described in greater detail below.
[0508] Symptoms of CoV infection may include fever, cough, runny nose, congestion, sore throat, bronchitis, pneumonia, shortness of breath, chest pain, headache, muscle ache, chills, fatigue, conjunctivitis, diarrhea, loss of smell, and loss of taste. Complications and/or diseases/disorders associated with coronavirus infection may include, for example, bronchitis, pneumonia, respiratory failure, acute respiratory failure, organ failure, multi-organ system failure, pediatric inflammatory multisystem syndrome, acute respiratory distress syndrome (a severe lung condition that causes low oxygen in the blood and organs), blood clots, cardiac conditions, myocardial injury, myocarditis, heart failure, cardiac arrest, acute myocardial infarction, dysrhythmias, venous thromboembolism, post intensive care syndrome, shock, anaphylactic shock, cytokine release syndrome, septic shock,
disseminated intravascular coagulation, ischemic stroke, intracerebral hemorrhage, microangiopathic thrombosis, psychosis, seizure, nonconvulsive status epilepticus, traumatic brain injury, stroke, anoxic brain injury, encephalitis, posterior reversible leukoencephalopathy, necrotizing encephalopathy, post- infectious encephalitis, autoimmune mediated encephalitis, acute disseminated encephalomyelitis, acute kidney injury, acute liver injury, pancreatic injury, immune thrombocytopenia, subacute thyroiditis, gastrointestinal complications, aspergillosis, increased susceptibility to infection with another virus or bacteria, and/or pregnancy-related complications. Certain diseases and conditions, such as high blood pressure, type 1 diabetes, liver disease, overweight, chronic lung diseases including cystic fibrosis, pulmonary fibrosis, and asthma, compromised immune system due to transplant, use of an immunosuppressant, or HIV infection, and brain and nervous system condition, may increase the risk of CoV infection-associated complications and diseases.
[0509] Also, the subject anti-CoV-S antibodies and antigen-binding fragments may be used alone or in conjunction with other active agents, e.g., opioids and non-opioid analgesics such as NSAIDs to elicit analgesia. In some embodiments, aspirin and/or acetaminophen may be taken in conjunction with the subject anti-CoV-S antibody or antigen-binding fragment. Aspirin is another type of non steroidal anti-inflammatory compound.
[0510] The subject antibodies potentially optionally may be combined with one or more of the following: (i) an antiviral drug, optionally, remdesivir, favipiravir, darunavir, nelfinavir, saquinavir, lopinavir, or ritonavir; (ii) an antihelminth drug, optionally ivermectin; (iii) an antiparasitic drug, optionally hydroxychloroquine, chloroquine, or atovaquone; (iv) antibacterial vaccine, optionally the tuberculosis vaccine BCG; or (v) an anti-inflammatory drug, optionally a steroid such as ciclesonide, a TNF inhibitor (e.g., adalimumab), a TNF receptor inhibitor (e.g., etanercept), an IL-6 inhibitor (e.g., clazakizumab), an IL-6 receptor inhibitor (e.g., toclizumab), or metamizole; (vi) an antihistamine drug, optionally bepotastine; (vii) an ACE inhibitor, which is optionally moexipril; or (viii) a drug that inhibits priming of CoV-S, optionally a serine protease inhibitor, further optionally nafamostat. in order to increase or enhance pain management. This may allow for such analgesic compounds to be administered for longer duration or at reduced dosages thereby potentially alleviating adverse side effects associated therewith.
[0511] The subject to which the pharmaceutical formulation is administered can be, e.g., any human or non-human animal needing such treatment, prevention and/or amelioration, or who would otherwise benefit from the inhibition or attenuation of CoV-S-mediated activity. For example, the subject can be an individual that is diagnosed with, or who is deemed to be at risk of being afflicted by any of the aforementioned diseases or disorders. In some instances the subject may be in an advanced state of CoV infection, e.g., a subject who is on a ventilator. In some instances, the subject can be one having one or more risk factors (such as advanced age, obesity, diabetes, etc, and others previously identified) which correlate to a poor CoV treatment or recovery prognosis. The present
disclosure further includes the use of any of the pharmaceutical formulations disclosed herein in the manufacture of a medicament for the treatment, prevention and/or amelioration of any disease or disorder associated with CoV or CoV-S activity (including any of the above-mentioned exemplary diseases, disorders and conditions).
[0512] The present invention also provides a method for treating a coronavirus infection in a subject in need thereof, the method comprising administering to the subject an isolated antibody, or antigen binding fragment thereof, wherein the antibody, or antigen-binding fragment thereof, comprises a heavy chain variable region (VH) comprising a VH CDR1 comprising SEQ ID NO:52, a VH CDR2 comprising SEQ ID NO:54, and a VH CDR3 comprising SEQ ID NO:56, and a light chain variable region (VL) comprising a VL CDR1 comprising SEQ ID NO:252, a VL CDR2 comprising SEQ ID NO:254, and a VL CDR3 comprising SEQ ID NO:256; and wherein the antibody, or antigen-binding fragment thereof, is administered at a dosage of at least 300 mg within 5, 4, 3, 2, or 1 days of symptom onset. In some embodiments, the antibody, or antigen-binding fragment thereof, is administered at a dosage of about 300 mg. In some embodiments, the antibody, or antigen-binding fragment thereof, is administered at a dosage of about 500 mg. In some embodiments, the antibody, or antigen-binding fragment thereof, is administered at a dosage of about 600 mg. In some embodiments, the antibody, or antigen-binding fragment thereof, is administered at a dosage of about 1200 mg. In some embodiments, the antibody, or antigen-binding fragment thereof, is administered at a dosage of about 4500 mg.
[0513] In some embodiments, the subject has at least one risk factor which renders them more prone to a poor clinical outcome. In some embodiments, the at least one risk factor is selected from the group consisting of: an old age selected from the group consisting of over 55, over 60 or over 65 years old; diabetes, a chronic respiratory condition, obesity, hypertension, a cardiac or cardiovascular condition, a chronic inflammatory or autoimmune condition, and an immune compromised status. In some embodiments, the subject is immunocompromised. In some embodiments, the subject is at a high risk of disease progression. In some embodiments, the subject is (a) age 50 years or above with no comorbid conditions or additional risk factors for progression of COVID-19; (b) between about 18 to about 50 years old and with one or more preexisting medical conditions selected from the group consisting of obesity, diabetes, chronic kidney disease, chronic lung disease, cardiac disease, sickle cell disease or thalassemia, solid organ or blood stem cell transplant recipient, other immunodeficiency due to underlying illness or immunosuppressant medication, Down Syndrome, stroke or cerebrovascular disease, substance use disorder, and pregnancy; or (c) between about 12 to about 17 years old and with one or more preexisting medical conditions selected from the group consisting of obesity, diabetes, chronic kidney disease, sickle cell disease or thalassemia, congenital or acquired heart disease, neurodevelopmental disorder, a medically-related technological dependence, asthma or chornic respiratory disease, solid organ or blood stem cell transplant recipient,
other immunodeficiency due to underlying illness or immunosuppressant medication, Down Syndrome, stroke or cerebrovascular disease, substance use disorder, and pregnancy. In some embodiments, the subject is age 50 or above with no comorbid conditions or additional risk factors for progression of COVID-19. In some embodiments, the subject has hypertension with at least one medication prescribed or recommended. In some embodiments, the subject has moderate to severe asthma requiring daily therapy.
J. Administration
[0514] In one embodiment, the anti-CoV-S antibodies described herein, or CoV-S binding fragments thereof, as well as combinations of said antibodies or antigen-binding fragments thereof, are administered to a subject at a concentration of between 0.1 mg/ml and about any one of 0.5, 1, 5, 10, 15 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 110, 120, 130, 140, 150, 160, 170,
180, 190, or 200 mg/ml, +/-10% error.
[0515] In another embodiment, the anti-CoV-S antibodies and fragments thereof described herein are administered to a subject at a dose of between about 0.01 and 100.0 or 200.0 mg/kg of body weight of the recipient subject. In certain embodiments, depending on the type and severity of the CoV-S- related disease, about 1 μg/kg to 50 mg/kg (e.g., 0.1-20 mg/kg) of antibody is an initial candidate dosage for administration to the patient, whether, for example, by one or more separate administrations, or by continuous infusion. In another embodiment, about 1 μg/kg to 15 mg/kg (e.g., 0.1 mg/kg- 10 mg/kg) of antibody is an initial candidate dosage for administration to the patient. A typical daily dosage might range from about 1 μg/kg to 100 mg/kg or more, depending on several factors, e.g., the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the agent, the method of administration, the scheduling of administration, and other factors known to medical practitioners. However, other dosage regimens may be useful.
[0516] For example, in addition to the relative dosages (mg/kg) discussed herein, the subject anti- CoV-S antibodies and antigen-binding fragments thereof can be administered to a subject at an absolute dose (mg). Accordingly, in one embodiment, the anti-CoV-S antibodies and antigen-binding fragments thereof described herein are administered to a subject at a dose of between about 1 microgram and about 2000 milligrams regardless of the route of administration.
[0517] In some embodiments, the antibody, or antigen-binding fragment thereof, is administered at a dose of about 100 mg to about 5000 mg, about 100 mg to about 4500 mg, about 100 mg to about 4000 mg, about 100 mg to about 3500 mg, about 100 mg to about 3000 mg, about 100 mg to about 2500 mg, about 300 mg to about 4500 mg, about 500 mg to about 4500 mg, about 600 mg to about 4500 mg, about 1200 mg to about 4500 mg, about 100 mg to about 2000 mg, about 200 mg to about 1500 mg, about 300 mg to about 600 mg, about 500 mg to about 1200 mg, or about 300 mg to about 1200
mg. In some embodiments, the antibody, or antigen-binding fragment thereof, is administered at a dose of about 150 mg, about 200 mg, about 300 mg, about 400 mg, about 450 mg, about 500 mg, about 600 mg, about 700 mg, about 800 mg, about 900 mg, about 1000 mg about 1100 mg, about 1200 mg, about 1300 mg, about 1400 mg, about 1500 mg, about 1600 mg, about 1700 mg, about 1800 mg, about 1900 mg, about 2000 mg, about 2500 mg, about 3000 mg, about 3500 mg, about 4000 mg, about 4500 mg, or about 5000 mg.
[0518] In some embodiments, the antibody, or antigen-binding fragment thereof, is administered intravenously. In other embodiments, the antibody, or antigen-binding fragment thereof, is administered intramuscularly.
[0519] In some embodiments, the antibody, or antigen-binding fragment thereof, is administered at a dose of about 150 mg intramuscularly, about 300 mg intramuscularly, about 450 mg intramuscularly, about 500 mg intravenously, about 600 mg intramuscularly, about 1200 mg intramuscularly, or about 1200 mg intravenously.
[0520] In some embodiments, at least one of the antibody, or antigen-binding fragment thereof, is administered. In some embodiments, at least two of the antibody, or antigen-binding fragment thereof, are administered. In some embodiments, the anti-CoV-S antibody and antigen-binding fragment thereof, e.g., ADI-58125, may be used in combination with a second antibody, or antigen-binding fragment thereof, wherein the second antibody, or antigen-binding fragment thereof, is selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130, ADI-58131, or a combination thereof. In some embodiment, the second antibody, or antigen-binding fragment thereof, is ADI-58122. In one embodiment, the second antibody, or antigen-binding fragment thereof, is ADI-58127. In one embodiment, the second antibody, or antigen-binding fragment thereof, is ADI-58129. In one embodiment, the second antibody, or antigen-binding fragment thereof, is ADI-58131.
[0521] In one embodiment, the antibody, or antigen-binding fragment thereof, is administered once. In one embodiment, the antibody, or antigen-binding fragment thereof, is administered weekly. In another embodiment, the antibody, or antigen-binding fragment thereof, is administered daily, weekly, every two weeks, monthly, or every two months. In one embodiment, the antibody, or antigen-binding fragment thereof, is administered weekly for about four weeks, once weekly for about a month, weekly for about 5 weeks, weekly for about 6 weeks, weekly for about 7 weeks, or weekly for about two months.
[0522] In some embodiments, the methods further comprise obtaining a serum sample from the subject.
[0523] In some embodiments, the antibody, or antigen-binding fragment thereof, reaches a maximum concentration (Cmax) of about 10 μg/mL to about 1000 μg/mL, about 20 μg/mL to about 500 μg/mL, about 30 μg/mL to about 400 μg/mL, about 40 μg/mL to about 300 μg/mL, about 50 μg/mL to about
200 mg/mL, about 50 mg/mL to about 100 mg/mL, about 30 mg/mL to about 70 mg/mL, about 100 mg.mL to about 400 mg/mL, or about 150 mg/mL to about 350 mg/mL in the serum sample of the subject. In some embodiments, the antibody, or antigen-binding fragment thereof, reaches a maximum concentration (Cmax) of about 30 mg/mL, about 40 mg/mL, about 50 mg/mL, about 60 mg/mL, about 70 mg/mL, about 80 mg/mL, about 90 mg/mL, about 100 mg/mL, about 110 mg/mL, about 120 mg/mL, about 130 mg/mL, about 140 mg/mL, about 150 mg/mL, about 160 mg/mL, about 170 mg/mL, about 180 mg/mL, about 190 mg/mL,or about 200 mg/mL, about 210 mg/mL, about 220 mg/mL, about 230 mg/mL, about 240 mg/mL, about 250 mg/mL, about 260 mg/mL, about 270 mg/mL, about 280 mg/mL, about 290 mg/mL, about 300 mg/mL, about 350 mg/mL, or about 400 mg/mL in the serum sample of the subject.
[0524] In some embodiments, the median time for the antibody, or antigen-binding fragment thereof, to reach a maximum concentration in the serum sample of the subject is about 5-30 days, about 6-20 days, about 7-18 days, or about 8-15 days, or about 13-15 days after administration. In some embodiments, the median time for the antibody, or antigen-binding fragment thereof, to reach a maximum concentration in the serum sample of the subject is about 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 days after administration.
[0525] In some embodiments, the antibody, or antigen-binding fragment thereof, reaches a maximum concentration (Cmax) of about 30 μg/mL to about 100 μg/mL, about 40 μg/mL to about 80 μg/mL, about 50 μg/mL to about 70 μg/mL, or about 30 μg/mL to about 65 μg/mL in the serum sample of the subject in about 6-20 days, about 7-18 days, about 8-15 or about 13-15 days after administration. [0526] In some embodiments, the area under the serum concentration-time curve from day 0 to day 21 (AUC0-21d) is about 100-2000 day* μg/mL, about 1000-2000 day* μg/mL, about 1400-2000 day* μg/mL, about 200-1500 day* μg/mL, about 400-1400 day* μg/mL, about 500-1300 day* μg/mL, about 600-1000 day* μg/mL, or about 800-900 day* μg/mL.
[0527] In some embodiments, the area under the serum concentration-time curve from day 0 to day 21 (AUC0-21d) is about 500 day* μg/mL, about 600 day* μg/mL, about 700 day* μg/mL, about 800 day* μg/mL, about 900 day* μg/mL, about 1000 day* μg/mL, about 1100 day* μg/mL, about 1200 day* μg/mL, about 1300 day* μg/mL, about 1400 day* μg/mL, about 1500 day* μg/mL, about 1600 day* μg/mL, about 1700 day* μg/mL, about 1800 day* μg/mL, about 1900 day* μg/mL, or about 2000 day* μg/mL.
[0528] In some embodiments, the area under the serum concentration-time curve from day 0 to day 90 (AUC0-90d) is about 1000-10000 day* μg/mL, about 2000-5000 day* μg/mL, about 3000-4000 day* μg/mL, about 5000-10000 day* μg/mL, about 5000-8000 day* μg/mL, or about 6000-8000 day* μg/mL.
[0529] In some embodiments, the area under the serum concentration-time curve from day 0 to day 90 (AUC0-90d) is about 1000 day* μg/mL, about 2000 day* μg/mL, about 3000 day* μg/mL, about
4000 day* mg/mL, about 5000 day* mg/mL, about 6000 day* mg/mL, about 7000 day* mg/mL, about 8000 day* mg/mL, about 9000 day* mg/mL, or about 10000 day* mg/mL.
[0530] In some embodiments, the area under the serum concentration-time curve from day 0 to day 180 (AUC0-180d) is about 1000-10000 day* mg/mL, about 2000-5000 day* mg/mL, about 3000-6000 day* mg/mL, about 5000-10000 day* mg/mL, about 5000-8000 day* mg/mL, or about 4000-6000 day* mg/mL.
[0531] In some embodiments, the area under the serum concentration-time curve from day 0 to day 180 (AUC0-180d) is about 1000 day* mg/mL, about 2000 day* mg/mL, about 3000 day* mg/mL, about 4000 day* mg/mL, about 5000 day* mg/mL, about 6000 day* mg/mL, about 7000 day* mg/mL, about 8000 day* mg/mL, about 9000 day* mg/mL, or about 10000 day* mg/mL.
[0532] In some embodiments, the area under the serum concentration-time curve from day 0 to day 365 (AUCo 365d) is about 1000-30000 day* mg/mL, about 5000-30000 day* mg/mL, about 10000- 30000 day* mg/mL, about 10000-15000 day* mg/mL, about 20000-30000 day* mg/mL, or about 25000-30000 day* mg/mL.
[0533] In some embodiments, the area under the serum concentration-time curve from day 0 to day 365 (AUCo 365d) is about 1000 day* mg/mL, about 2000 day* mg/mL, about 3000 day* mg/mL, about 4000 day* mg/mL, about 5000 day* mg/mL, about 6000 day* mg/mL, about 7000 day* mg/mL, about 8000 day* mg/mL, about 9000 day* mg/mL, about 10000 day* mg/mL, about 11000 day* mg/mL, about 12000 day* mg/mL, about 13000 day* mg/mL, about 14000 day* mg/mL, about 15000 day* mg/mL, about 20000 day* mg/mL, about 21000 day* mg/mL, about 22000 day* mg/mL, about 23000 day* mg/mL, about 24000 day* mg/mL, about 25000 day* mg/mL, about 26000 day* mg/mL, about 27000 day* mg/mL, about 28000 day* mg/mL, about 29000 day* mg/mL, or about 30000 day* mg/mL. [0534] In some embodiments, the antibody, or antigen-binding fragment thereof, has a serum half- life of about 50-200 days, about 50-160 days, about 50-140 days, about 40-130 days, about 50-100 days, about 60-100 days, about 60-90 days, about 70-90 days, about 90-100 days, about 70-80 days, about 80-130 days, about 100-150 days, about 110-140 days, about 120-140 days. In some embodiments, the antibody, or antigen-binding fragment thereof, has a serum half-life of about 50, 55, 60, 65, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117, 118,
119, 120, 121, 122, 123, 124, 125, 126, 127, 128, 129, 130, 131, 132, 133, 134, 135, 136, 137, 138, 139, 140, 141, 142, 143, 144, 145, 146, 147, 148, 149, 150, 151, 152, 153, 154, 155, 156, 157, 158, 159, 160, 165, 170, 75, 180, 185, 190, 195, or 200 days.
[0535] In some embodiments, the method further comprises obtaining an epithelial lining fluid (ELF) sample from the subject. The ELF sample may comprise an ELF sample from the upper airway, e.g., nasopharyngeal or oropharyngeal; the lower airway, e.g., lung, and/or an alveolar tissue.
[0536] In some embodiments, the antibody, or antigen-binding fragment thereof, reaches a concentration of about 1 mg/mL to about 100 mg/mL, about 1 mg/mL to about 80 mg/mL, about 80 mg/mL to about 100 mg/mL, about 50 mg/mL to about 100 mg/mL, about 1 mg/mL to about 50 mg/mL, about 2 mg/mL to about 25 mg/mL, or about 2 mg/mL to about 10 mg/mL in the ELF sample of the subject.
[0537] In some embodiments, the antibody, or antigen-binding ragment thereof, has a receptor occupancy of at least 50%, 60%, 70%, 80%, or 90% in the ELF sample. Receptor occupany (RO) can be linked to a viral dynamic model to enable the prediction of the natural time course of viral load and the effect of the antibody, or antigen-binding fragment thereof, on viral clearance and infectivity rate. In some embodiments, RO can be calculated using: 1) in vitro SARS-CoV-2 binding kinetics of the antibody, or antigen-binding fragment thereof, e.g., the association rate constant (kon) and the dissociation rate constant (k0ff) , e.g., obtained from a Biacore assay; 2) time course of the concentrations of the antibody, or antigen-binding fragment thereof, in a sample, e.g., an ELF sample; and 3) time course of viral load following administration of the antibody, or antigen-binding fragment thereof.
[0538] In some embodiments, administration of the antibody, or antigen-binding fragment thereof, results in at least 50%, 60%, 70%, 80%, or 90% SARS-CoV-2 receptor occupancy in the ELF sample. In some embodiments, administration of the antibody, or antigen-binding fragment thereof, results in a durable, e.g., about 20-day, about 25-day, about 28-day, about 30-day, about 40-day, about 50-day, about 60-day, SARS-CoV-2 receptor occupancy in the ELF sample. In other embodiments, administration of the antibody, or antigen-binding fragment thereof, maintains the concentration of the antibody, or antigen binding fragment thereof, above a concentration (e.g., about 0.27 mg/L or about 0.5 mg/L) associated with about 95% or 100% viral growth suppression in an in vitro post infection assay.
[0539] In some embodiments, intramuscular (IM) administration of ADI-58125 at a dose of about 300 mg results in about 90% SARS-CoV-2 receptor occupacy in the ELF sample for at least 28 days, and maintains the concentration of ADI-58125 in the ELF sample above the concentration associated with 100% viral growth suppression in vitro, i.e., 0.5 mg/mL.
[0540] In some embodiments, intramuscular (IM) administration of ADI-58125 at a dose of about 300 mg results in about 90% SARS-CoV-2 receptor occupacy in the ELF sample for at least 28 days, and maintains the concentration of ADI-58125 in the ELF sample above the concentration associated with 95% viral growth suppression in vitro, i.e., 0.274 mg/mL.
[0541] In some embodiments, administration of the antibody, or antigen-binding fragment thereof, results in a 50% virus neutralization titer (MN50) of about 100-2000, e.g., within the range of peak serum virus-neutralizing antibody (sVNA) titers for COVID-19 vaccine recipients. In some embodiments, administration of the antibody, or antigen-binding fragment thereof, maintains a serum
concentration of the antibody, or antigen-binding fragment thereof, about 100-fold higher than the in vitro 90% inhibitory concentration (IC90) of 0.011 μg/mL against authentic SARS-CoV-2 (USA- WA1) for a minimum of 6 months.
[0542] In some embodiments, the virus neutralization titer of the antibody, or antigen-binding fragment thereof, is determined using a plaque reduction neutralization test (PRNT), as described in Example 1. The PRNT is a serological test which utilizes the ability of a specific antibody to neutralize a virus, in turn, preventing the virus from causing the formation of plaques in a cell monolayer (Stephen J. et al., The American Journal of Tropical Medicine and Hygiene. 81 (5): 825- 833). Typically, the assay involves mixing a constant amount of virus with dilutions of the serum specimen being tested, followed by plating of the mixture onto cells of an appropriate cell line for the individual virus. The concentration of plaque forming units can be determined by the number of plaques formed after a few days. A vital dye (e.g., neutral red) is then added for visualization of the plaques and the number of plaques in an individual plate is divided by the original number of virons to calculate the percentage neutralization. Depending on the virus, the plaque forming units are measured by microscopic observations, fluorescent antibodies, or specific dyes that react with the infected cell. Currently, the PRNT test is considered the "gold standard" for detecting and measuring antibodies that can neutralize the viruses that cause many diseases. It has a higher sensitivity and is more specific than many other serological tests for the diagnosis of some viruses. In some embodiments, the 80% neutralization titer is reported using a validated method, e.g., SOP-VC-M204. In some embodiments, the 50% neutralization titer is reported.
[0543] In some embodiments, the antibody, or antigen-binding fragment thereof, has a virus neutralizing titer of about 100-2000, about 200-1500, about 300-1500, or about 500-1500 in the serum sample of the subject about 3, 6, or 12 months after administration. In some embodiments, the antibody, or antigen-binding fragment thereof, has a virus neutralizing titer of about 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, 1100, 1200, 1300, 1400, 1500, 1600, 1700, 1800, 1900 or 2000 in the serum sample of the subject about 3, 6 or 12 months after administration.
[0544] In some embodiments, the 80% virus neutralization titer (MN80) of the antibody, or antigen binding fragment thereof, at about days 0-14, about days 7-21, about 3 months, or about 6 months after administration, is about 10-6000, about 50-600, about 500-1500, about 1000-2500, about 100- 2500, about 500-2000, about 500-1500, about 400-1200, about 200-1500, about 300-1000, about 400- 800, about 400-1000, or about 500-600 in the subject.
[0545] In some embodiments, the 80% virus neutralization titer (MN80) of the antibody, or antigen binding fragment thereof, at about day 7, day 14, or day 21 after administration, is about 300-2000, about 400-600, about 600-1500, about 1100-1700, about 500-1700, about 500-1500, about 400-1200, about 400-800, about 400-1000, or about 500-600 in the serum sample of the subject.
[0546] In some embodiments, the 80% virus neutralization titer (MN80) of the antibody, or antigen binding fragment thereof, at about 3 months after administration, is about 200-1000, about 200-800, about 200-500, about 400-900, or about 400-600 in the serum sample of the subject.
[0547] In some embodiments, the 80% virus neutralization titer (MN80) of the antibody, or antigen binding fragment thereof, at about 6 months after administration, is about 10-500, about 300-500, or about 50-200 in the serum sample of the subject.
[0548] In some embodiments, the 50% virus neutralization titer (MN50) of the antibody, or antigen binding fragment thereof, at about days 0-14, about days 7-21, about 3 months, about 6 months, or about 12 months after administration, is about 100-6000, about 300-1500, about 1700-3800, about 3800-5200, about 300-5500, about 1200-4500, about 1300-4300, about 1200-4000, about 100-2500, about 500-2500, about 800-2000, about 1000-1800, about 800-1300, about 900-1100, or about 1300- 1500 in the serum sample of the subject.
[0549] In some embodiments, the 50% virus neutralization titer (MN50) of the antibody, or antigen binding fragment thereof, at about day 7, day 14 or day 21 after administration, is about 1000-4500, about 1200-4500, about 1300-4300, about 1200-3900, about 1500-4000, about 1800-3800, about 3800-4500, about 1000-1800 or about 1200-1500 in the serum sample of the subject.
[0550] In some embodiments, the 50% virus neutralization titer (MN50) of the antibody, or antigen binding fragment thereof, at about 3 months after administration, is about 800-1300 or about 900- 1100 in the serum sample of the subject. In some embodiments, the 50% virus neutralization titer (MN50) of the antibody, or antigen-binding fragment thereof, at about 6 months after administration, is about 200-500 or about 300-600 in the serum sample of the subject.
[0551] In some embodiments, the 50% virus neutralization titer (MN50) of the antibody, or antigen binding fragment thereof, at about 12 months after administration, is about 150-500 or about 200-400 in the serum sample of the subject.
[0552] In some embodiments, the serum concentration of the antibody, or antigen-binding fragment thereof, is about 1-300 mg/L, about 1-250 mg/L, about 1-200 mg/L, about 1-100 mg/L, about 100-250 mg/L, about 150-200 mg/L, about 120-170 mg/L, about 1-90 mg/L, about 1-80 mg/L, about 1-70 mg/L, about 1-60 mg/L, about 1-50 mg/L, about 1-40 mg/L, about 1-30 mg/L, about 1-20 mg/L, about 1-10 mg/L, about 2-9 mg/L, about 3-8 mg/L, about 4-6 mg/L, about 5-80 mg/L, about 10-70 mg/L, or about 10-60 mg/L in the subject about 3 months after administration.
[0553] In some embodiments, the serum concentration of the antibody, or antigen-binding fragment thereof, is about 1 mg/L, about 2 mg/L, about 3 mg/L, about 4 mg/L, about 5 mg/L, about 6 mg/L, about 7 mg/L, about 8 mg/L, about 9 mg/L, about 10 mg/L, about 11 mg/L, about 12 mg/L, about 13 mg/L, about 14 mg/L, about 15 mg/L, about 16 mg/L, about 17 mg/L, about 18 mg/L, about 19 mg/L, about 20 mg/L, about 25 mg/L, about 30 mg/L, about 35 mg/L, about 40 mg/L, about 45 mg/L, about 50 mg/L, about 55 mg/L, about 60 mg/L, about 65 mg/L, about 70 mg/L, about 75 mg/L, about 80
mg/L, about 85 mg/L, about 90 mg/L, about 95 mg/L, about 100 mg/L, about 110 mg/L, about 120 mg/L, about 130 mg/L, about 140 mg/L, about 150 mg/L, about 160 mg/L, about 170 mg/L, about 180 mg/L, about 190 mg/L, about 200 mg/L, about 210 mg/L, about 220 mg/L, about 230 mg/L, about 240 mg/L, about 250 mg/L, about 300 mg/L in the subject about 3 months after administration. [0554] In some embodiments, the serum concentration of the antibody, or antigen-binding fragment thereof, is about 1-100 mg/L, about 1-90 mg/L, about 1-80 mg/L, about 1-70 mg/L, about 1-60 mg/L, about 1-50 mg/L, about 1-40 mg/L, about 1-30 mg/L, about 1-20 mg/L, about 1-10 mg/L, about 2-9 mg/L, about 3-8 mg/L, about 4-6 mg/L, about 5-80 mg/L, about 10-70 mg/L, about 10-60 mg/L, about 5-25 mg/L, about 10-30 mg/L, about 15-25 mg/L, about 20-100 mg/L, about 30-100 mg/L, about 50- 80 mg/L, or about 60-70 mg/L in the subject about 6 months after administration.
[0555] In some embodiments, the serum concentration of the antibody, or antigen-binding fragment thereof, is about 1 mg/L, about 2 mg/L, about 3 mg/L, about 4 mg/L, about 5 mg/L, about 6 mg/L, about 7 mg/L, about 8 mg/L, about 9 mg/L, about 10 mg/L, about 11 mg/L, about 12 mg/L, about 13 mg/L, about 14 mg/L, about 15 mg/L, about 16 mg/L, about 17 mg/L, about 18 mg/L, about 19 mg/L, about 20 mg/L, about 25 mg/L, about 30 mg/L, about 35 mg/L, about 40 mg/L, about 45 mg/L, about 50 mg/L, about 55 mg/L, about 60 mg/L, about 65 mg/L, about 70 mg/L, about 75 mg/L, about 80 mg/L, about 85 mg/L, about 90 mg/L, about 95 mg/L, or about 100 mg/L in the subject about 6 months after administration.
[0556] In some embodiments, the serum concentration of the antibody, or antigen-binding fragment thereof, is about 0.1-30 mg/L, about 1-30 mg/L, about 1-20 mg/L, about 1-10 mg/L, about 10-20 mg/L, about 0.1-3 mg/L, about 0.5-8 mg/L, about 0.5-10 mg/L, about 2-9 mg/L, about 5-15 mg/L, about 3-8 mg/L, or about 4-6 mg/L in the subject about 12 months after administration.
[0557] In some embodiments, the serum concentration of the antibody, or antigen-binding fragment thereof, is about 0.1 mg/L, about 0.2 mg/L, about 0.3 mg/L, about 0.4 mg/L, about 0.5 mg/L, about 0.6 mg/L, about 0.7 mg/L, about 0.8 mg/L, about 0.9 mg/L, about 1 mg/L, about 2 mg/L, about 3 mg/L, about 4 mg/L, about 5 mg/L, about 6 mg/L, about 7 mg/L, about 8 mg/L, about 9 mg/L, about 10 mg/L, about 15 mg/L, about 20 mg/L, about 25 mg/L, or about 30 mg/L in the subject about 12 months after administration.
[0558] In some embodiments, the clearance rate for the antibody, or antigen-binding fragment thereof, is about 0.1-10 mL/d, about 0.1-5.0 mL/d, about 0.1-3.0 mL/d, about 0.5-2.5 mL/d, or about 1.0-2.0 mL/d.
[0559] In some embodiments, the steady state volume of distribution for the antibody, or antigen binding fragment thereof, is about 1-10 L, about 2-8 L, about 4-9 L, about 4-8 L, or about 5-7L.
[0560] In some embodiments, administration of the antibody, or antigen-binding fragment thereof, reduces the risk of COVID-19 hospitalization or death of the subject. In some embodiments, the risk
of COVID-19 hospitalization or death is reduced by at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90% or 95%.
[0561] In some embodiments, administration of the antibody, or antigen-binding fragment thereof, reduces viral load in the subject. In some embodiments, the viral load is reduced by at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90% or 95%. In some embodiments, the viral load is reduced by at least about 0.5 loglO copies/mL, at least about 0.6 loglO copies/mL, at least about 0.7 loglO copies/mL, at least about 0.8 loglO copies/mL, at least about 0.9 loglO copies/mL, at least about 1.0 loglO copies/mL, at least about 1.1 loglO copies/mL, at least about 1.2 loglO copies/mL, at least about 1.3 loglO copies/mL, at least about 1.4 loglO copies/mL, at least about 1.5 loglO copies/mL, at least about 1.6 loglO copies/mL, at least about 1.7 loglO copies/mL, at least about 1.8 loglO copies/mL, at least about 1.9 loglO copies/mL, at least about 2.0 loglO copies/mL, at least about 2.1 loglO copies/mL, at least about 2.2 loglO copies/mL, at least about 2.3 loglO copies/mL, at least about 2.4 loglO copies/mL, at least about 2.5 loglO copies/mL, at least about 2.6 loglO copies/mL, at least about 2.7 loglO copies/mL, at least about 2.8 loglO copies/mL, at least about 2.9 loglO copies/mL, or at least about 3.0 loglO copies/mL.
[0562] In a preferred embodiment, the anti-CoV-S antibodies described herein, or anti-CoV-S antigen-binding fragments thereof, as well as combinations of said antibodies or antigen-binding fragments thereof, are administered to a recipient subject with a frequency of once every twenty-six weeks or less, such as once every sixteen weeks or less, once every eight weeks or less, once every four weeks or less, once every two weeks or less, once every week or less, or once daily or less. [0563] According to preferred embodiments, the antibody containing medicament or pharmaceutical composition is peripherally administered to a subject via a route selected from one or more of: orally, sublingually, buccally, topically, rectally, via inhalation, transdermally, subcutaneously, intravenously, intra-arterially, or intramuscularly, via intracardiac administration, intraosseously, intradermally, intraperitoneally, transmucosally, vaginally, intravitreally, epicutaneously, intra- articularly, peri-articularly, or locally.
[0564] Fab fragments may be administered every two weeks or less, every week or less, once daily or less, multiple times per day, and/or every few hours. In one embodiment, a patient receives Fab fragments of 0.1 mg/kg to 40 mg/kg per day given in divided doses of 1 to 6 times a day, or in a continuous perfusion form, effective to obtain desired results.
[0565] It is to be understood that the concentration of the antibody or Fab administered to a given patient may be greater or lower than the exemplary administration concentrations set forth above. [0566] A person of skill in the art would be able to determine an effective dosage and frequency of administration through routine experimentation, for example guided by the disclosure herein and the teachings in, Goodman & Gilman's The Pharmacological Basis of Therapeutics, Brunton, L.L. et al. editors, 11th edition, New York, New York: McGraw-Hill (2006); Howland, R. D. et al,
Pharmacology, Volume 864, Lippincott's illustrated reviews., Philadelphia, PA: Lippincott Williams & Wilkins (2006); and Golan, D. E., Principles of pharmacology: the pathophysiologic basis of drug therapy, Philadelphia, PA: Lippincott Williams & Wilkins (2007).
[0567] In another embodiment, the anti-CoV-S antibodies described herein, or CoV-S binding fragments thereof, as well as combinations of said antibodies or antigen-binding fragments thereof, are administered to a subject in a pharmaceutical formulation. In a preferred embodiment, the subject is a human.
[0568] In some embodiments, the subject is an adult. In some embodiment, the subject is an adolescent. In some embodiments, the subject is a child or a pediatric subject. In one embodiment, the subject is a pediatric subject, e.g., from birth to age of 11 years old, e.g., birth to < 1 month, 1 month to <2 years, or 2-11 years old. In other embodiments, the adolescent is 12-17 years old or 12- 15 years old. In one embodiment, the adult is over the age of 18. In some embodiments, the subject has no known recent exposure to COVID-19. In some embodiments, the subject has a known recent exposure to COVID-19. In one embodiment, recent exposure is within the last 14 days, within the last 10 days, within the last 7 days, within the last 6 days, 5 days, 4 days, 3 days, 2 days, or 24 hours. In some embodiments, the subject has COVID-19 symptoms or an active COVID-19 infection. In some embodiments, the subject has had COVID-19 symptoms for less than 5 days, less than 4 days, less than 3 days, less than 2 days, or less than 1 day. In some embodiments, the subject is treated within 5 days, 4 days, 3 days, 2 days or 1 day of symptom onset. In some embodiments, early treatment, e.g., receiving treatment within 5 days, 4, days, 3 days, 2 days or 1 day of symptom onset, provides a reduction in the risk of COVID-19 related hospitalization and all-cause death. In some embodiments, the subject, e.g., the adult, the adolescent, or the child, is vaccinated or has received a COVID-19 vacination. In some embodiments, the subject, e.g., the adult, the adolescent, or the child, is not vaccinated or has not received a COVID-19 vacination. In some embodiments, the subject, e.g., the adult, the adolescent, or the child, is at risk of COVID-19 exposure. For example, the subject may be a first responder, doctor, nurse, clinican, etc.
[0569] In some embodiments, the subject is immunocompromised. In some embodiments, the subject is a significantly immune compromised subject, e.g., adult subject or pediatric subject, but whose underlying medical condition(s) or treatments to treat those conditions cause to the subjects to be immunocompromised, and/or put them at increased risk of developing severe symptomatic COVID- 19. In some embodiments, the subject has no known recent exposure to COVID-19. In some embodiments, the subject has a known recent exposure to COVID-19. In one embodiment, recent exposure is within the last 14 days, within the last 10 days, within the last 7 days, within the last 6 days, 5 days, 4 days, 3 days, 2 days, or 24 hours. In some embodiments, the immunocomprised subject includes, but is not limited to, an individual who is actively treated for solid tumor or hematologic malignancies; solid organ transplant (SOT) recipient taking immunosuppressive therapy;
CAR-T-cell therapy or HCT recipient; subjects having moderate or severe primary immunodeficiency; subjects having advanced or untreated HIV infection, or who are taking high-dose corticosteroids, alkylating agents, antimetabolites, or TNF blockers. In some embodiments, the immunocompromised subject is vaccinated or has received a COVID-19 vacination. In some embodiments, the immunocompromised subject is not vaccinated or has not received a COVID-19 vacination.
[0570] In some embodiments, the subject is at a high risk of COVID progression. In some embodiments, the subjects at a high risk of disease progression are age 50 years old or above with no comorbid conditions or additional risk factors for progression of COVID-19.
[0571] In some embodiments, the subjects at a high risk of disease progression are those about 18 to about 50 years old and with one or more preexisting medical conditons selected from the group consisting of (i) obesity (body mass index (BMI) > 30 kg/m2), (ii) diabetes (type 1 or type 2); (iii) chronic kidney disease (eGFR calculated by modification of diet in renal disease (MDRD) of 59 mL/min/1.73 m2 or less, including end-stage renal disease on hemodialysis); (iv) chronic lung disease (emphysema/chronic obstructive pulmonary disease, chronic bronchitis, interstitial lung disease [including idiopathic pulmonary fibrosis], cystic fibrosis, or moderate to severe asthma [defined as requiring daily therapy]); (v) cardiac disease (heart failure, coronary artery disease, cardiomyopathies, or hypertension [with at least one medication prescribed or recommended]); (vi) sickle cell disease or thalassemia; (vii) solid organ or blood stem cell transplant recipients; (viii) other immunodeficiency due to underlying illness or immunosuppressant medication (e.g., corticosteroids >20 mg/day prednisone or equivalent); (ix) Down Syndrome, (x) stroke or cerebrovascular disease, which affects blood flow to the brain; (xi) substance use disorder; or (xii) pregnant.
[0572] In some embodiments, the subjects at a high risk of disease progression are those about 12 to about 17 years old (inclusive) and with one or more preexisting medical conditions selected from the group consisting of (i) BMI >85th percentile for age and sex based on United States Center for Disease Control (CDC) growth charts; (ii) diabetes (Type 1 or Type 2); (iii) chronic kidney disease; (iv) sickle cell disease or thalassemia; (v) congenital or acquired heart disease; (vi) neurodevelopmental disorders (e.g., cerebral palsy, Down syndrome); (vii) a medically-related technological dependence (e.g., tracheostomy, gastrostomy, or positive pressure ventilation not related to COVID-19); (viii) asthma, reactive airway or other chronic respiratory disease that requires daily medication for control; (ix) solid organ or blood stem cell transplant recipients; (x) other immunodeficiency due to underlying illness or immunosuppressant medication; (xi) substance use disorder; or (xii) pregnant.
[0573] In some embodiments, the subject is age 50 or above with no comorbid conditions or additional risk factors for progression of COVID-19.
[0574] In some embodiments, the subject has hypertension with at least one medication prescribed or recommended.
[0575] In some embodiments, the subject has moderate to severe asthma requiring daily therapy. [0576] A “pharmaceutical composition” or “medicament” refers to a chemical or biological composition suitable for administration to a subject, preferably a mammal, more preferably a human. Such compositions may be specifically formulated for administration via one or more of a number of routes, including but not limited to buccal, epicutaneous, epidural, inhalation, intraarterial, intracardial, intracerebroventricular, intradermal, intramuscular, intranasal, intraocular, intraperitoneal, intraspinal, intrathecal, intravenous, oral, parenteral, rectally via an enema or suppository, subcutaneous, subdermal, sublingual, transdermal, and transmucosal. In addition, administration can occur by means of injection, powder, liquid, gel, drops, or other means of administration.
[0577] In one embodiment, the anti-CoV-S antibodies or antigen-binding fragments thereof, as well as combinations of said antibodies or antigen-binding fragments thereof, may be optionally administered in combination with one or more active agents. Such active agents include (i) an antiviral drug, optionally, remdesivir, favipiravir, darunavir, nelfinavir, saquinavir, lopinavir, or ritonavir; (ii) an antihelminth drug, optionally ivermectin; (iii) an antiparasitic drug, optionally hydroxychloroquine, chloroquine, or atovaquone; (iv) antibacterial vaccine, optionally the tuberculosis vaccine BCG; or (v) an anti-inflammatory drug, optionally a steroid such as ciclesonide, a TNF inhibitor (e.g., adalimumab), a TNF receptor inhibitor (e.g., etanercept), an IL-6 inhibitor (e.g., clazakizumab), an IL-6 receptor inhibitor (e.g., toclizumab), or metamizole; (vi) an antihistamine drug, optionally bepotastine; (vii) an ACE inhibitor, optionally moexipril; or (viii) a drug that inhibits priming of CoV-S, optionally a serine protease inhibitor, further optionally nafamostat.
[0578] An anti -histamine can be any compound that opposes the action of histamine or its release from cells (e.g., mast cells). Anti-histamines include but are not limited to acrivastine, astemizole, azatadine, azelastine, betatastine, brompheniramine, buclizine, cetirizine, cetirizine analogues, chlorpheniramine, clemastine, CS 560, cyproheptadine, desloratadine, dexchlorpheniramine, ebastine, epinastine, fexofenadine, HSR 609, hydroxyzine, levocabastine, loratadine, methscopolamine, mizolastine, norastemizole, phenindamine, promethazine, pyrilamine, terfenadine, and tranilast.
[0579] In CoV infection, respiratory symptoms are often exacerbated by additional bacterial infection. Therefore, such active agents may also be antibiotics, which include but are not limited to amikacin, aminoglycosides, amoxicillin, ampicillin, ansamycins, arsphenamine, azithromycin, azlocillin, aztreonam, bacitracin, carbacephem, carbapenems, carbenicillin, cefaclor, cefadroxil, cefalexin, cefalothin, cefalotin, cefamandole, cefazolin, cefdinir, cefditoren, cefepime, cefixime, cefoperazone, cefotaxime, cefoxitin, cefpodoxime, cefprozil, ceftazidime, ceftibuten, ceftizoxime, ceftobiprole, ceftriaxone, cefuroxime, cephalosporins, chloramphenicol, cilastatin, ciprofloxacin,
clarithromycin, clindamycin, cloxacillin, colistin, co-trimoxazole, dalfopristin, demeclocycline, dicloxacillin, dirithromycin, doripenem, doxycycline, enoxacin, ertapenem, erythromycin, ethambutol, flucloxacillin, fosfomycin, furazolidone, fusidic acid, gatifloxacin, geldanamycin, gentamicin, gly copeptides, herbimycin, imipenem, isoniazid, kanamycin, levofloxacin, lincomycin, linezolid, lomefloxacin, loracarbef, macrolides, mafenide, meropenem, methicillin, metronidazole, mezlocillin, minocycline, monobactams, moxifloxacin, mupirocin, nafcillin, neomycin, netilmicin, nitrofurantoin, norfloxacin, ofloxacin, oxacillin, oxytetracycline, paromomycin, penicillin, penicillins, piperacillin, platensimycin, polymyxin B, polypeptides, prontosil, pyrazinamide, quinolones, quinupristin, rifampicin, rifampin, roxithromycin, spectinomycin, streptomycin, sulfacetamide, sulfamethizole, sulfanilamide, sulfasalazine, sulfisoxazole, sulfonamides, teicoplanin, telithromycin, tetracycline, tetracyclines, ticarcillin, tinidazole, tobramycin, trimethoprim, trimethoprim- sulfamethoxazole, troleandomycin, trovafloxacin, and vancomycin.
[0580] Active agents also include aldosterone, beclomethasone, betamethasone, corticosteroids, cortisol, cortisone acetate, deoxycorticosterone acetate, dexamethasone, fludrocortisone acetate, glucocorticoids, hydrocortisone, methylprednisolone, prednisolone, prednisone, steroids, and triamcinolone. Any suitable combination of these active agents is also contemplated.
[0581] A “pharmaceutical excipient” or a “pharmaceutically acceptable excipient” is a carrier, usually a liquid, in which an active therapeutic agent is formulated. In one embodiment, the active therapeutic agent is a humanized antibody described herein, or one or more fragments thereof. The excipient generally does not provide any pharmacological activity to the formulation, though it may provide chemical and/or biological stability, and release characteristics. Exemplary formulations can be found, for example, in Remington’s Pharmaceutical Sciences, Gennaro, A. editor, 19th edition, Philadelphia, PA: Williams and Wilkins (1995), which is incorporated by reference.
[0582] As used herein “pharmaceutically acceptable carrier” or “excipient” includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic, and absorption delaying agents that are physiologically compatible. In one embodiment, the carrier is suitable for parenteral administration. Alternatively, the carrier can be suitable for intravenous, intraperitoneal, intramuscular, or sublingual administration. Pharmaceutically acceptable carriers include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active compound, use thereof in the pharmaceutical compositions of the disclosure is contemplated. Supplementary active compounds can also be incorporated into the compositions. [0583] Pharmaceutical compositions typically must be sterile and stable under the conditions of manufacture and storage. The disclosure contemplates that the pharmaceutical composition is present in lyophilized form. The composition can be formulated as a solution, microemulsion, liposome, or
other ordered structure suitable to high drug concentration. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol), and suitable mixtures thereof. The disclosure further contemplates the inclusion of a stabilizer in the pharmaceutical composition. The proper fluidity can be maintained, for example, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
[0584] In many cases, it will be preferable to include isotonic agents, for example, sugars, polyalcohols such as mannitol and sorbitol, or sodium chloride in the composition. Absorption of the injectable compositions can be prolonged by including an agent that delays absorption, for example, monostearate salts and gelatin. Moreover, the alkaline polypeptide can be formulated in a time-release formulation, for example in a composition that includes a slow release polymer. The active compounds can be prepared with carriers that will protect the compound against rapid release, such as a controlled release formulation, including implants and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, poly anhydrides, polyglycolic acid, collagen, polyorthoesters, polylactic acid, polylactic and polyglycolic copolymers (“PLG”). Many methods for the preparation of such formulations are known to those skilled in the art. [0585] For each of the recited embodiments, the compounds can be administered by a variety of dosage forms. Any biologically acceptable dosage form known to persons of ordinary skill in the art, and combinations thereof, are contemplated. Examples of such dosage forms include, without limitation, reconstitutable powders, elixirs, liquids, solutions, suspensions, emulsions, powders, granules, particles, microparticles, dispersible granules, cachets, inhalants, aerosol inhalants, patches, particle inhalants, implants, depot implants, injectables (including subcutaneous, intramuscular, intravenous, and intradermal), infusions, and combinations thereof.
[0586] The above description of various illustrated embodiments of the disclosure is not intended to be exhaustive or to limit the disclosure to the precise form disclosed. While specific embodiments of, and examples for, the disclosure are described herein for illustrative purposes, various equivalent modifications are possible within the scope of the disclosure, as those skilled in the relevant art will recognize. The teachings provided herein of the disclosure can be applied to other purposes, other than the examples described herein.
[0587] Certain anti-CoV-S antibody polynucleotides and polypeptides are disclosed in the sequence listing accompanying this patent application filing, and the disclosure of said sequence listing is herein incorporated by reference in its entirety.
[0588] The entire disclosure of each document cited (including patents, patent applications, journal articles, abstracts, manuals, books, or other disclosures) in the Background, Detailed Description, and Examples is herein incorporated by reference in their entireties.
[0589] The following examples are put forth so as to provide those of ordinary skill in the art with a complete disclosure and description of how to make and use the subject disclosure and are not intended to limit the scope of what is regarded as the invention. Efforts have been made to ensure accuracy with respect to the numbers used (e.g. amounts, temperature, concentrations, etc.), but some experimental errors and deviations should be allowed for. Unless otherwise indicated, parts are parts by weight, molecular weight is average molecular weight, temperature is in degrees centigrade; and pressure is at or near atmospheric.
EXAMPLES
Example 1: Phase 1, Randomized, Double-blind, Single Ascending Dose Study [0590] This Example provides safety, pharmacokinetics, and virus neutralizing antibody titer data of ADI-58125 in a Phase 1, randomized, double-blinded, single ascending dose study. In this Phase 1 study, three dose levels of ADI-58125: 300 mg IM, 500 mg IV, and 600 mg IM, were assessed in healthy participants. All dose levels utilized a 100 mg/ml concentration of ADI-58125.
[0591] Biefly, subjects were randomized 8 active:2 placebo, and each dose level had a sentinel cohort of 1 active: 1 placebo. To accommodate differences in anticipated Tmax, IM and IV subjects were confined to the Phase 1 unit for 72 and 24 hours after dosing, respectively. Subjects in the IM cohorts were assessed for injection site reactions daily through discharge. Safety monitoring was ongoing, with weekly phone calls and clinic visits at 3, 6, 9, and 12 months after randomization. Demographics
[0592] Thirty healthy volunteers received ADI-58125 or placebo in this study. Baseline characteristics were well balanced among cohorts (Table 10). The average age of participants was 37.3 (range: 18 - 55) years and 53% (16) are female; 40% (12) participants identified as Hispanic or Latino. One subject identified as American Indian or Alaska Native, three participants identified as Black or African American, one participant identified as Asian, two participants identified as Native Hawaiian or Other Pacific Islander, and 24 participants identified as White. Participants were permitted to select more than one race. The average BMI of participants was 24.0 (range: 19.9 - 29.4) kg/m2. All subjects received the full dose of study medication and all remain in the study.
Safety and Tolerability
[0593] No serious adverse events, adverse events of special interest, study drug-related adverse events, discontinuations, deaths, or injection site reactions, or hypersensitivity reactions were observed.
[0594] Cohort 1 includes 300 mg IM ADI-58125 (N=8) and placebo (N=2) participants. One participant from Cohort 1 (300 mg IM) reported a single episode of dizziness, which occurred shortly after IM injection and resolved after the participant was permitted to eat. This AE was graded mild, deemed unrelated to study medication, and resolved without concomitant medication administration (see, Tables 11 and 12).
[0595] Cohort 2 includes 500 mg IV ADI-58125 (N=8) and placebo (N=2) participants. One participant in Cohort 2 (500 mg IV) reported musculoskeletal chest pain beginning approximately 10 hours after infusion and resolving prior to discharge the following morning. No abnormalities were found on physical exam or on an ECG performed during the event. This event was graded mild, deemed unrelated to study medication, and resolved without intervention or concomitant medication administration.
[0596] A second participant in Cohort 2 (500 mg IV) reported a single episode of vomiting. This event occurred approximately 24 hours after infusion and resolved immediately after the event and prior to discharge. No abnormalities were found on physical exam shortly after the event. This event was graded mild, deemed unrelated to study medication, and resolved without intervention or concomitant medication administration.
[0597] Cohort 3 includes 600 mg IM ADI-58125 (N=8) and placebo (N=2) participants. One participant in Cohort 3 (600 mg IM) reported bruising on their left thigh, which they noticed incidentally 11 days after their injection and reported at her Day 14 visit. This finding was not noted during injection site monitoring, which included physical exams, nor was it noted or reported at their Day 7 visit. This event was graded mild, deemed unrelated to study medication.
[0598] All available adverse event, laboratory, physical exam and vital sign data for Cohort 1 were evaluated and it was concluded that the single adverse event in Cohort 1 was unrelated to study drug.
No clinically relevant laboratory, physical exam or vital sign values were found during the review. No other concerns were identified.
Table 11. Summary of Adverse Events (Solicited and Unsolicited), Safety Population - Cohort 1 through Day 2.
0599] AE= Adverse event, TEAE = Treatment emergent adverse event, SAE = Serious adverse event. N is number of participants in the Safety Population. Percentages are based on the total number of participants in Safety [0600] Population.
[0601] [1] Only study procedure AEs were collected from the time that informed consent was obtained until study drug dosing.
[0602] [2] TEAE is defined as any event not present prior to the initiation of study drug or any event already present that worsens in either intensity or frequency following exposure to study drug.
[0603] [3] Solicited AEs are recorded through Day 4 following study drug administration. Local reactions after Day 4 are considered non-solicited AEs. Missing relationship to study drug is imputed as “Related” and all solicited AEs are assumed to be “Related”. Missing severity is imputed as “Severe”. A Grade 4 (potentially life threatening) solicited AE is considered a SAE.
[0604] For participant counts, if participants experienced one or more events, they are counted only once. For AE counts, each occurrence of an AE is counted. AEs are summarized at maximum Severity and strongest Relationship to study drug.
[0605] Adverse event terms are coded using MedDRA version 23.1.
Table 12. Summary of Treatment-Emergent Adverse Events (Solicited and Unsolicited) by System Organ Class, Preferred Term and Severity, Safety Population - Cohort 1 through Day 21.
[0606] TEAE = Treatment emergent adverse event. N is number of participants in the Safety Population. Percentages are based on the total number of participants in Safety Population.
[0607] At each level of summarization (SOC or PT), participants who experienced more than one TEAE were only counted once at maximum Severity.
[0608] Missing severity of study adverse event is imputed as “Severe”.
[0609] Solicited AEs are local reactions to the IM injection and are collected through Study Day 4 and are imputed as related to study drug.
[0610] TEAEs is defined as any event not present prior to the initiation of study drug or any event already present that worsens in either intensity or frequency following exposure to study drug.
[0611] Adverse event terms are coded using MedDRA version 23.1.
[0612] Through a minimum of 12 weeks post dose, 11 AEs were reported in 7 participants (2/10 [20%] Cohort 1; 3/10 [30%] Cohort 2; 2/10 [20%] Cohort 3); all were mild in severity and considered unrelated to study drug.
Pharmacoki neti cs
[0613] Pharmacokinetics (PK) of 300 mg IM dose were evaluated using blood samples collected prior to dosing (predose) and at 8 h, 24 h (Day 2), Day 3, Day 4, Day 7, Day 14, and Day 21 postdose. ADI-58125 serum concentrations were measured using a validated LB A (ligand binding assay)-hybrid LC -MS/MS assay at Q2 Solutions (Ithaca, NY) with a quantitative range of 0.5 μg/mL to 200 μg/mL. Blinding to treatment was maintained for study team personnel. Only external testing labs (Q2 Solutions and Viroclinics) and pharmacology vendors (Certara and ePD/ICPD) were unblinded to individual participant treatment.
[0614] Preliminary interim pharmacokinetic analysis demonstrated observable serum concentrations in 8 subjects following a single IM administration of 300 mg ADI-58125 (Figure 1). Maximum concentration (Cmax), median time to maximum concentration (Tmax), and area under the serum concentration-time curve from 0 to 20 days (AUCo 20d) are summarized in Table 13. Based upon these data, the quantitative systems pharmacology/physiologically based pharmacokinetic (QSP/PBPK) model predicted the observed ADI-58125 concentrations well and with relatively little bias across the 21 -day sampling interval (Figure 2). An approximate 80-day ADI-58125 terminal elimination half- life was predicted by the QSP/PBPK model.
[0615] Consistent with the model predicted half-life, further pharmacokinetic analysis also demonstrated that ADI-58125 had a potential for prolonged protection. As shown in Figure 17, a half-life of about 60-100 days was observed for a single IM adminsitration of 300 mg ADI-58125 (n=8).
[0616] Safety, tolerability and pharmacokinetics (PK) were also assessed 3 months and 6 months post dose. Through 3 months post dose, 8 adverse events (AEs), mild in severity, were reported; no
study drug-related AEs, serious AEs, injection-site reactions, or hypersensitivity reactions were reported. The PK profile through Day 180 for a single 300 mg IM injection of ADI-58125 and through Day 90 for 500 mg IV and 600 mg IM doses of ADI-58125 was dose proportional and consistent with an extended-half-life mAh (Figure 19).
[0617] The median time to maximum concentration (Tmax) was 13 days (range: 6-20) after a single 300 mg IM injection.
[0618] Based on the 6-month data cohecte from participants who received 300 mg IM, the estimated half-life of ADI-58125 was 96.8 days.
Includes only participants who received ADI-58125.
Abbreviations: AUC = area under the serum concentration-time curve, Cmax = maximum concentration, SD = standard deviation, Tmax = time to maximum concentration, T1/2, half life.
Neutralizing Antibodies
[0619] Virus neutralizing antibody (VNA) titers generated in the context of prior infection or vaccination have been shown to be a key correlate of protection from COVID-19 (Khoury DS, et al, , medRxiv. 2021 Jan 1; Earle KA, et al, medRxiv. 2021 Jan 1). These data provide a useful supportive marker for evaluating the proposed dose of ADI-58125 for use in the setting of prevention.
[0620] The peak 50% SARS-CoV-2 neutralizing geometric mean titer (GMT) of 437 generated two weeks post second dose of the mRNA vaccine BNT162b2 (Mulligan MJ, et al, Nature. 2020 Oct;586(7830):589-593) was originally used to evaluate the proposed ADI-58125 dose of 300 mg IM, with the goal of maintaining VNA near this threshold. However, recent analysis of 6-month follow-up data from the pivotal trial for BNT162b2 demonstrated vaccine efficacy of 91.3% through 6 months, when VNA titers are likely to be much lower than the peak observed two weeks after the second dose. Although neutralizing antibody titer data are not available for the corresponding population for which vaccine efficacy was reported, other studies have shown an approximately 10-fold decrease in neutralizing antibody titers through Day 150 for subjects vaccinated with BNT162b2 (Wang Z, et al,
Nature, 2021 Feb 10: 1-7). Similar decay in neutralizing antibody titer profiles have been demonstrated for convalescent sera (Wheatley AK, et al, Nat Commun. 2021 Feb 19; 12(1): 1162) and neutralizing antibody responses induced by the Moderna mRNA vaccine (Widge et al, N Engl J Med. 2021 Jan 7;384(l):80-82; Wang et al, Nature. 2021 Feb 10:1-7). For example, peak live virus ID50 geometric mean VNA titers for the Moderna vaccine mRNA- 1273 were -1000 for all age groups tested at Day 43 post vaccination 1 and decayed to 406, 171 and 131 by Day 209 in participants aged 18 to 55, 56 to 70 and 71 years of age or older, respectively (Doria-Rose N, et al, N Engl J Med. 2021 Apr 6). Taken together, these data support the idea that lower VNA titers are protective against symptomatic disease.
[0621] Serum virus neutralizing titers were evaluated using blood samples collected prior to dosing (predose) and at 8 h, 24 h (Day 2), Day 7, and Day 14 postdose. A plaque reduction neutralization test (PRNT) run at Viroclinics Biosciences (Rotterdam, NL) was used to measure serum titers against SARS-CoV-2 variant BavPat-1/2020 (with D614G). Virus was incubated with serial dilutions of serum, and virus/serum mixtures were then added to a confluent monolayer of Vero-E6 cells. Infected areas were immunostained 16-24 hours after infection and quantified using automated detection. The 80% neutralization titer (MN80) is reported using a validated method (SOP-VC-M204); the 50% neutralization titer (MN50) is a nonvalidated readout and reported for informational purposes.
[0622] Analysis of serum neutralizing titers from participants receiving a single 300 mg IM dose of ADI-58125 was presented in Figure 3. By day 13 post-injection, 80% neutralization titers [MN80, geometric mean (CV%)] were 569 (43.8%) and 50% neutralization titers [MN50, geometric mean (CV%)] were 1382 (32.7%). Samples from participants receiving placebo demonstrated no consistent response above the lower limit of detection (LLOD).
[0623] A linear regression model was fit to the time-matched individual MN50 titers and ADI-58125 concentrations demonstrated a strong correlation between the two measures (R2=0.955). Linear interpolation of the QSP/PBPK model-predicted median ADI-58125 concentration at 6 months post injection (8.03 μg/mL) predicts a median MN50 titer of approximately 247.
[0624] This value is likely within the range of 50% SARS-CoV-2 neutralizing GMT titers remaining -6 months post vaccination for mRNA vaccines such as BNT162b2 and mRNA-1273 (Wang et al, Nature. 2021 Feb 10:1-7; Doria-Rose N, et al., N Engl J Med. 2021 Apr 6).
[0625] Serum viral neutralizing antibody (sVNA) titers were also assessed Day 2, Day 7, Day 14, Day 21, 3 months, and 6 months post dose in subjects following a single IM administration of 300 mg ADI-58125, a single IM administration of 600 mg ADI-58125, and a single IV administration of 500 mg ADI-58125. The sVNA data are presented in Table 14. ADI-58125 sVNA titers against authentic SARS-CoV-2 were compared with AZD1222- and mRNA-1273-induced titers measured by plaque reduction neutralization assay. ADI-58125 serum concentration was highly predictive of the MN50 sVNA titer (Figure 20).
Table 14. Neutralizing Antibody Titers by Study Day and Treatment Groups
Abbreviations: IM = intramuscular, IV = intravenous, MN50 = 50% neutralization titer, MN80 = 80% neutralization titer.
Values shown as geometric mean (coefficient of variation %).
[0626] After a single 300 mg IM dose of ADI-58125, 50% sVNA titers (geometric mean) were 346 on Day 2, 1380 on Day 14, and were maintained through Day 21. By Day 2, MN50 sVNA titers significantly exceeded peak titers associated with AZD1222 and were similar to peak titers associated with mRNA-1273 (Figure 18). By Day 7, MN50 sVNA titers associated with ADI-58125 were significantly higher than pean titers associated with AZD1222 (P<0.0001) and mRNA-1273 (P<0.01), and were maintained at comparable levels to titers associated with mRNA-1273 through Study Month 6 (Figure 18), suggesting that the ADI-58125 neutralizing antibody titers also support potential for prolonged protection.
[0627] sVNA titers following ADI-58125 administration were also determined by plaque reduction assay for SARS-CoV-2 BavPat (D614G), B.1.351/Beta and the B.1.617.2/Delta variants. These were compared with titers 7-30 days after the second dose of authorized vaccines in a separate cohort of volunteers. As shown in Figure 27, 80% neutralization (MN80) sVNA titres following a single 300 mg IM injection of ADI-58125 exceeded titers against each of the BavPat, B.1.351/Beta and B.1.617.2/Delta variants 7-30 days following 2-dose mRNA-1273 vaccination, and this trend was maintained for at least 3 months. By 6 months, ADI-58125 sVNA titres were 0.6-, 1.4-, and 1.8-fold of peak titers induced by mRNA-1273 vaccination for variants tested. Statistical summary of MN80 comparisons following ADI-58125 administration and 2-dose mRNA 1273 vaccination was shown in Table below. P-values were determined by two-sided Mann Whitney U test. Thus, these results further support the potential for a single 300 mg IM injection of ADI-58125 to provide prolonged protection against symptomatic COVID-19 for a minimum of 6 months.
Conclusions
[0628] In summary, no safety or tolerability issues were observed in any of the dose cohorts, including the 300 mg IM dose and two higher doses to establish a safety margin (500 mg IV and 600 mg IM). Thus, a single dose of ADI-58125, up to 600 mg IM, was well tolerated. Preliminary interim data indicate that ADI-58125 PK following IM administration of a single 300 mg dose was consistent with the QSP/PBPK model predictions. These PK data and the sVNA titer data support the potential for a single 300 mg IM injection of ADI-58225 to treat COVID-19 and provide prolonged protection from COVID-19 for a minimum of 6 months.
[0629] Taken altogether, the absence of safety and tolerability signals, PK results that confirm previous model predictions, and adequate early VNA titers provided in this Example support further evaluation of a single 300 mg IM injection of ADI-58125 in the ongoing Phase 2/3 trials for the prevention and treatment of COVID-19.
Example 2: Serum Titers of ADI-58125 Against SARS-CoV-2 Variants by Plaque Reduction Neutralization Test
[0630] Plaque reduction neutralization test (PRNT) was used to measure serum titers against SARS- CoV-2 D614G and B.1.351 variants. Briefly, 400 PFU SARS-CoV-2 virus was incubated with serial dilutions of ADI-58125 for 1 hour. Virus/antibody mixtures were then added to a confluent monolayer of Vero-E6 cells and incubated for 48 hours before adding neutral red agar overlay. Plaques were counted 24 hours after infection and the concentration of antibody that reduced the plaques by 50% (PRNT50) or 90% (PRNT90) relative to control (i.e., virus alone with no antibody) was calculated (Figure 4). The neutralizing antibody titers to reduce the ability of both virus variants from infecting host cells by 50% or 90% were shown in the table below.
Example 3: Post-Infection Assay with ADI-58125 Against SARS-CoV-2 Variants [0631] A post-infection assay was performed to measure the concentration of antibody required to prevent viral replication of the SARS-CoV-2 D614G and B.1.351 variants in infected cells. Briefly, cells were infected with the SARS-CoV-2 D614G or B.1.351 variants. After 1 hour incubation, unbound virus was washed away, and medium containing various concentration of ADI-58125 antibody was added to infected-cell monolayers. Viral supernatant was sampled daily and infectious viral burden was quantified by plaque assay on Vero E6 cells (Figure 5).
[0632] The concentration curves showing the amount of antibody required to prevent viral replication in infected cells were shown in Figure 6. As shown in Figure 6, 0.125 μg/ml ADI-58125 was able to reduce the viral burden for both variants when compared to the control, and the viral burden decreased significantly as the concentration of the antibody increased.
Example 4: Potent Neutralizing Activity of ADI-58125 and ADI-58122 Against a Broad Panel of Variants
[0633] Neutralizing activity of ADI-58125 and ADI-58122 against naturally circulating SARS-CoV- 2 variants with single and/or double point mutations and emerging lineages including the Alpha/B.1.1.7 varaint, the Beta/B.1.351 variant, the Gamma/P.l variant, the B.1.617 variants (e.g.,
B.1.617.1/Kappa and B.1.617.2/Delta), the Omicron lineages BA.l., BA.1.1, and BA.2, the
C.37/Lambda variant, and the Mu/B.1621 variant were assessed. Several other SARS-CoV-2 antibodies were included as comparators. Briefly, neutralizing activity against authentic SARS-CoV-2 virus varaints was assessed in a focus reduction neutralization test using Vero cells. Half-maximal inhibitory concentration (IC50) values were determined using nonlinear regression curve fitting.
Several SARS-CoV-2 variants containing single or double amino acid mutations in the spike protein and full sets of mutations observed in variants of interest and variants of concern (VOCs) were assessed in a lentivial pseudovirus assay. D614G, an early variant of SARS-CoV-2, was used as a reference to calculate the IC50 fold change in neutralizing activity of the antibodies. In vitro neutralizing activity of ADI-58125 against SARS-CoV-2 variants, including those reported to exhibit reduced susceptibility to emergency-use authorized (EUA) mAbs, was assessed in a non-replicative vesicular stomatitis virus pseudovirus assay. Fold reduction was calculated by dividing the IC50 of a variant by the mean IC50 (14.6 ng/mL) of the D614G reference strain.
[0634] As shown in Figure 7, Figure 8A and Figure 8B, ADI-58125 and ADI-58122 were able to neutralize all common circulating SARS-CoV-2 variants and emerging lineages. In particular, ADI- 58125 retained in vitro neutralizing activity within 0.4- to 5.1-fold relative to the reference D614G variant for all pseudovirus variants tested, including the newly emerging lineages including the Alpha/B.1.1.7 variant, the Beta/B.1.351 variant, the Gamma/P.l variant, the B.1.617 (e.g.,
B.1.617.1/Kappa and B.1.617.2/Delta), the Delta plus/AY.l, the C.37/Lambda, and the Mu/B.1621
variants (Figure 8A) and variants with single -/double-spike mutations (Figure 8B). Furthremore, ADI-58125 maintained neutralizing activity against all tested SARS-CoV-2 variants incorporating mutations reported to confer increased resistance to EUA m Abs (Figure 8C). IC50 values for ADI- 58125 (within 0.6- to 2.4-fold of the D614G strain) were lower than those observed for bamlanivimab, etesevimab, casirivimab, imdevimab, and sotrovimab. These data support further clinical investigation of ADI-58125 not only for COVID-19 but also for potential utility in future outbreaks caused by other SARS-like viruses.
Example 5: A Whole-Body Quantitative System Pharmacology Physiologically-Based Pharmacokinetic (QSP/PBPK) Model that a priori Predicts Intramuscular (IM) Pharmacokinetics of ADI-58125: an Extended Half-life Monoclonal Antibody for the Treatment and Prevention of Coronavirus Disease ( COVID-19 )
[0635] This Example provides a QSP/PBPK model, which was constructed using ADI-58125- specific physiochemical properties and published non-human primate (NHP) and human PK data for other antibodies, to a priori predict and confirm NHP and human PK.
[0636] An existing QSP/PBPK model was modified to include 3 distinct lung sub-compartments: upper airway, lower airway, and alveolar tissue (Figure 9A). Each sub-compartment (Figure 9B) contained an epithelial lining fluid (ELF) space (Figure 9B). The model was fit separately to digitized NHP and human serum PK data for 7 extended half-life antibodies to estimate the apparent neonatal Fc receptor (FcRn) binding affinity
and bioavailability by drug. Nasopharyngeal swab (upper airway) and lung (lower airway) ELF PK data from 4 additional antibodies were used to optimize a single rate constant for transcytosis in lung. Patches of positive charge was a covariate on the rate of pinocytosis of antibody entry and exit from the endosomal space (Figure 9B). Observed NHP (ADI- 58125 10 mg/kg IM) and human (ADI-58125 300 mg IM) PK data collected over the initial 21 days post dose were compared with model forecasts from a 1000-iteration simulation.
[0637] The distribution of fitted NHP KD,FcRn provided accurate predictions of NHP serum PK data (Figure 9C). NHP ADI-58125 KD,FcRn was optimized to be 35.7 nM and human ADI-58125 KD,FcRn (9.55 nM) was derived using a mean NHP:human KD,FcRn ratio of 3.74 across antibodies. Model-based simulated human serum PK data using inter-subject variability from NHP and actual weight distribution from an ongoing Phase 1 study aligned with initial 21 -day data (Figure 9D). Using an adult CDC weight distribution (45-150 kg), the simulated median half-life was 74.4 days.
[0638] For initial QSP/PBPK model projections, the modified QSP whole-body PBPK model, estimated BHP and human apparent KD,FcRn distributions, an a reference US Centers for Disease Control body weight distribution were used to provide initial stimulation (1000 iterations) forecast of NHP and human ADI-58125 serum concentration-time profiles. When measured NHP ADI-58125
serum concentration became available, the raw data were overlaid on the initial median and 90% prediction interval (PI) forecast for NHPs.
[0639] For optimized QSP/PBPK model projections, the modified QSP/PBPK model was optimized by estimating NHP intramuscular (IM) bioavailability and ADI-58125 KD,FcRn and applying an NHP:human KD,FcRn ratio to the NHP KD,FcRn values estimated for ADI-58125 to better forecast human ADI-58125 concentration-time profiles. When measured human ADI-58125 serum concentration became available, the raw data were overlaid on the forecasted median and 90% prediction interval (PI) forecast for human. The QSP/PBPK model was then optimized by estimating KD,FcRn and IM availability using the interim human PK data, along with estimating inter-individual variability for some key parameters to better reflect observed variability.
[0640] Histograms of simulated human body weight and KD,FcRn distributions in humans and NHPs are shown in Figures 21A-21C.
[0641] Figures 22A-22B show the initial QSP/PBPK model-forecasted NHP median (90% prediction interval (PI)) serum ADI-58125 concentration-time profile following a single intravenous (IV) and intramuscular (IM) administration of 10 mg/kg ADI-58125 with measured concentration data overlaid. Figures 23A-23B show the optimized QSP/PBPK model-forecasted NHP median (90% PI) serum ADI-58125 concentration-time profile following a single intravenous (IV) and intramuscular (IM) administration of 10 mg/kg ADI-58125 with measured concentration data overlaid. The QSP whole -body PBPK model was optimized by estimating KD,FcRn (4.27 nM) and IM bioavailability (92%) using the interim human PK data, along with estimating inter-individual variability for some key parameters to better reflect observed variability. Figures 24A-24D show the observed and optimized QSP/PBPK model-forecasted human median (90% PI) serum ADI-58125 concentration-time profiles in healthy adult participants following a 300 mg IM, 500 mg IM or 500 mg IV administration of ADI-58125 with measured concentration data overlaid.
[0642] The modeling strategy involved the modification of a platform whole-body PBPK model designed for wild-type IgGl mAbs to forecast the PK of an extended half-life mAh. The modified QSP/PBPK model account for the altered binding affinity to FcRn and included patches of positive charge (PPC) as a covariate on the rate of pinocytosis into the endosomal space (CLup). The model adequately a priori predicted the observed ADI-58125 PK in NHPs and human, thus supporting the selected dose. This innovative QSP-based modeling and simulation approach enabled the evaluation of candidate dose regimens prior to the availability of PK data, supporting the rapid advancement of the ADI-58125 clinical program during the COVID-19 pandemic.
Example 6: A Whole-Body Quantitative System Pharmacology Physiologically-Based Pharmacokinetic (QSP/PBPK) Model to Support Dose Selection of ADI-58125 for the Treatment of Coronavirus Disease (COVID-19)
[0643] This Example provides a QSP/PBPK model to support dose selection for a Phase 2/3 COVID- 19 treatment study of ambulatory patients with mild to moderate COVID-19 (STAMP: NCT04805671).
[0644] The QSP/PBPK model comprised 15 specific tissues and one representing the rest of the body; each tissue was connected through blood and lymph flow to the systemic circulation. In tissue endothelial spaces, mAbs enter by pinocytosis (CLup) and via the interaction with neonatal Fc receptor (FcRn), FcRn-bound mAh is recycled, and unbound drug is eliminated (Kdeg). The QSP/PBPK model was modified such that the lung compartment was subdivided into alveoli and upper (naso- /oropharyngeal) and lower lung airway compartments (bronchi; Figure 9A). It was assumed that the upper, lower, and alveoli sub-compartments contribute 8.5%, 8.5%, and 83.0% toward lung volume and 2.5%, 5.0%, and 92.5% toward lung blood flow, respectively. It was assumed that FcRn concentrations in each lung sub-compartment were the same, mAbs recycle back to EFF and interstitial compartments, CFupin lung epithelium was 10-times slower than vascular endothelial cells, and the endosomal space was 0.5% of cellular space for pulmonary epithelial cells. mAbs were also allowed to cross epithelial cells by transcytosis (ktrans) and, upon entering the interstitial space, to either exit the lung via lymph flow or re-ente the vascular space via FcRn-mediated recycling ktrans was calibrated using serum PK data along with EFF and nasopharyngeal swab PK data from reference mAbs MHAA4549A, VIS-410, ASN-1, and ASN-2. It was assumed that binding to SARS-CoV-2 virus does not impact the ADI-58125 PK at clinically relevant doses.
[0645] A QSP/PBPK model was used to simulate receptor occupancy (RO) and drug exposure in the upper airway (nasopharyngeal/oropharyngeal epithelial lining fluid [EFF] compartment). RO was linked to an existing viral dynamic model to enable the prediction of the natural time course of viral load and the effect of ADI-58125 on viral clearance and infectivity rate.
[0646] RO was calculated using: 1) in vitro ADI-58125-SARS-CoV-2 binding kinetics (association rate constant (kon) of 1.52E+06 M -1•s-1 and dissociation rate constant (k0ff) of 2.81E-04 s-1 from a Biacore assay; 2) time course of QSP/PBPK model-forecasted ADI-58125 concentrations in upper aiway EFF of lung; and 3) time course of viral load following ADI-58125 administration at peak viral load ( e.g ., log 107 to log 109 copies/mF for all patients) using in vitro 50% half maximal concentration (IC50) of 0.007 μg/mF against authentic SARS-CoV-2 (e.g., Delta variant). Molar SARS-CoV-2 viral binding site capacity was calculated assuming 40 spike proteins per virion, 3 binding sites per spike, for all patients.
[0647] The QSP/PBPK model and a 2018 CDC reference body weight distribution (45-150 kg) were used to simulate 1000 concentration-time profiles for a range of candidate ADI-58125 regimens. ADI-
58125 regimens were evaluated against 2 criteria: 1) ability to attain near complete (>90%), and durable (28-day) SARS-CoV-2 RO in the ELF; and 2) ability to maintain ELF ADI-58125 concentrations >0.27 mg/L, which was associated with 95% viral growth suppression in an in vitro post-infection assay against the Beta variant.
[0648] A single 300 mg IM ADI-58125 dose met the dose selection criteria in terms of RO (Figure 10A) and viral growth suppression (Figure 10B). Figure 10A shows the QSP model-predicted median (90% prediction interval, PI) ADI-58125 PK profiles associated with viral growth suppression. Times to attaining relevant target concentrations are provided below.
[0649] Table 15 shows ADI-58125 potency against SARS-CoV-2 variants of concern.
[0650] As shown in Figure 10A and Table 15, ADGI-58125 attained ELF concentration above 10 times the IC90 of other variants of concern.
[0651] Figure 10B shows the ADI-58125 receptor occupancy at various Delta variant virion densities after a single 300 mg IM injection of ADI-58125.
[0652] These data support the evaluation of an ADI-58125 300 mg IM dose for the treatment of COVID-19. ADI-58125 is forecasted to attain near complete (>90%) SARS-CoV-2 RO in the ELF and maintain ELF ADI-58125 concentrations above that associated with 95% viral growth suppression in vitro.
[0653] In conclusion, the 300 mg IM regimen has a projected ability to rapidly achieve and maintain target tissue concentrations at key tissue sites of viral replication, including the ability to attain near complete (>90%) and durable (28-day) SARS-Cov-2 across a range of baseline viral loads.
Example 7: A Whole-Body Quantitative System Pharmacology Physiologically-Based Pharmacokinetic (QSP/PBPK) Model to Support Dose Selection of ADI-58125 for the Prevention of Coronavirus Disease (COVID-19)
[0654] This Example provides a QSP/PBPK model to support dose selection for a Phase 2/3 COVID- 19 prevention study (EVADE: NCT04859517).
[0655] Using the QSP/PBPK model and a CDC reference body weight distribution (45-150 kg),
1000 concentration-time profiles were simulated for a range of candidate ADI-58125 single injection regimens.
[0656] Prior to availabity of human PK data, the QSP PBPK model forecasts in humans were based upon the estimated IM bioavilability from NHP, while the KD,FcRn value of 9.55 nM was derived based upon multiplying the mean NHP:human KD,FcRn ratio for other extended half-life antibodies to the NHP KD,FcRn value for ADI-581258. The QSP/PBPK model was later optimized by estimating KD,FcRn (4.27 nM) and IM bioavailability (92.2%) using the interim human PK data, along with estimating inter-individual variability for some key parameters to better reflect observed variability.
[0657] ADI-58125 IM dosing regimens were evaluated against two criteria: (1) the ability to attain measured 50% neutralization (MN50) serum virus-neutralizing antibody (sVNA) titers within the range of peak sVNA titers for COVID-19 vaccine recipients; and (2) the ability to maintain serum ADI-58125 concentrations 100-fold higher than the in vitro 90% inhibitory concentration (IC90) of 0.011 μg/mL against authentic SARS-CoV-2 (USA-WA1) for a minimum of 6 months in >90% of simulated patients. This threshold was based on a precedent with respiratory syncytial virus and HIV, in which serum concentrations approximately 100-fold higher than the in vitro IC50 were associated with protection in animal models and/or in humans.
[0658] After measured human ADI-58125 serum concentrations became available, the QSP/PBPK model was optimized by refitting to the human data, allowing for formal estimation of interindividual variability, and ADI-58125 dose regimen simulations were updated.
[0659] Histograms of the simulated human body weight and KD,FcRn distributions in humans are shown in Figures 21A-B.
[0660] Using the original model, single ADI-58125 IM injections of 300 mg or greater were projected to maintain serum coencentration in most simulated patients for up to 12 months (Figures 25A-C). Table 16A shows the ADI-58125 potency against SARS-CoV-2 variants of concern.
[0661] The optimized model confirmed these predictions and suggests that the single 300 mg IM injection provides a margin of coverage of SARS-CoV-2 variants with high IC90 values than those of the original variant used to support this target (Figures 26A-C).
[0662] Based on the data from the first-in-human Phase 1 study (see Example 1), ADI-58125 maintains MN50 titers within the range of those achieved by COVID-19 vaccine recipients following 2 doses (AZD1222, mean titer 80: mRNA-1273, mean titer 327). Given a QSP/PBPK projected ADI-
58125 52-week post-dose medium serum concentration of 5.3 mg/L and a regression relating ADI- 58125 concentration and MN50 titer, the predicted MN50 is 231 one year post-dose.
[0663] These data support the evaluation of a single 300 mg IM injection of ADI-58125 for the prevention of COVID-19 in both preOand post-exposure settings. The single 300 mg IM regimen has a projected ability to rapid exceed the IC90 target in the majority of simulated patients, to maintain effective concentrations for up to 12 months, and to provide greater efficacy margins than lower disease for coverage against SARS-CoV-2 variants.
Example 8: In vivo Prophylactic Efficacy of ADI-58125 in Syrian Hamster [0664] The prophylactic efficacy of ADI-58125 was assessed in vivo in hamster. Briefly, Syrian hamsters were dosed intraperitoneally with a range of ADI-58125 doses (n=40; 9.25 - 2000 μg) or control mAh (sham-treated isotype matched IgG) (n=20; either 9.25 or 2000 μg) 24 hours prior to intranasal challenge with le5 plaque-forming units (pfu) of SARS-2/WA-1 to evaluate the prophylactic efficacy of ADI-58125 (Figure 11).
[0665] Hamseters were weighed daily over 6 days, while antibody titers, viral load and lung histopathology were assessed fowlloing sacrifice of five animals from each group on Day 3 and 6 post-challenge.
[0666] As shown in Figure 12A, an ADI-58125 dose of > 55 μg was associated with protection from weight loss compared with controls at Day 6. Hamsters receiving about 333 and 2000 μg doses displayed limited histopathological evidence of pneumonia (Figure 12B). Hamsters receiving the highest dose (2000 μg) had no detectable virus in lung samples (Figure 13). Specifically, treatment with 333 μg and 2000 μg of ADI-58125 significantly reduced infectious viral loads at Day 3 post exposure compared with controls (P<0.005). Similar trends were observed for viral sub-genomic RNA (sgRNA), which was produced during active viral infection. In addition, no evidence of antibody-dependent enhancement was observed across all ADI-58125 doses, including subtherapeutic doses.
[0667] In sum, these data demonstrated that prophylactic administration of ADI-58125 provides dose-dependent protection from SARS-CoV-2 infection in the Hamster model, and ADI-58125 conferred significant protection from weight loss and lung pathology and inhibited viral replication at the 333 and 2000 μg doses versus control antibody.
Example 9: In vivo Prophylactic Efficacy of ADI-58125 in Non Human Primates [0668] The prophylactic efficacy of ADI-58125 was further assessed in vivo in non human primates (NHP). Briefly, rhesus macaques were dosed intravenously with ADI-58125 (n=8; 5 or 25 mg/kg), or control mAh (sham-treated isotype matched IgG) (n=4; 25 mg/kg) 3 days prior to intranasal / intratracheal challenge with le6 pfu of SARS-2/WA-1 to evaluate prophylactic efficacy of ADI- 58125 (Figure 14).
[0669] Viral load was assessed in nasopharyngeal (NP) and oropharyngeal (OP) samples (daily), and in bronchoalveolar lavage (BAL) samples (Days 1, 3, and 5) by reverse transcription-polymerase chain reaction (RT-PCR, all samples) and plaque assays (NP and OP swabs only).
[0670] As shown in Figure 15, an ADI-58125 dose of 25 mg/kg was associated with reduced viral replication in the upper and lower airways with sgRNA below the limit of detection in all respiratory compartments tested. Specifically, a similar trend was observed for genomic RNA data (data not shown). Substantial protection was also observed at the 5 mg/kg dose level, as demonstrated by accelerated clearance of infectious virus. In addition, no viral particles were detected in NP and OP compartments at the 25 mg/kg ADI-58125 dose (Figure 16).
[0671] These data demonstrated that ADI-58125 confers potent protection from SARS-CoV-2 infection at dose ranges from 5-25 mg/kg in a NHP model. These results support further investigation of ADI-58125 for the prevention of COVID-19 in humans.
Example 10: A Phase 1/2, Open-Label, Comparator-Controlled Study Evaluating the Safety, Tolerability, and SARS-CoV-2 Antibody Titers of ADI-58125 Supplementation vs. BNT162b2 COVID-19 Vaccine Boost in Previously Vaccinated Adults
[0672] A Phase 1/2, randomized, open label, comparator-controlled, study is conducted in adults with no known history of Coronavirus Disease 19 (COVID-19) or Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection and who are a minimum of 6 months out from completion of a primary BNT162b2 COVID-19 vaccine series at the time of study drug dosing. Participants receive a single open-label IM injection of ADI-58125 or BNT162b2 vaccine boost and will be followed for 12 months.
[0673] Participants per cohort receive study drug (ADI-58125 or BNT162b2). Adult male and female participants must be age >18 years at the time of consent. Participants who are healthy or have stable chronic medical conditions, and who are a minimum of 6 months out from completion of a primary
COVID-19 vaccine series at the time of study drug dosing are enrolled and screened for participation within 28 days before the scheduled administration of study drug.
[0674] Screening procedures commence after written informed consent is obtained and include confirmation of inclusion and exclusion criteria, medical history, documentation of prior COVID-19 vaccination and medications (i.e., medications taken within 30 days before the scheduled dose of study drug), viral RT-PCR and serology tests, baseline laboratory testing including serum virus neutralizing antibody titers, vital sign measurements, and physical examination.
[0675] Key inclusion criteria include male or female subjects age >18 years at the time of consent; healthy or with stable chronic medical conditions, including immunocompromise, defined as disease not requiring significant change in therapy or hospitalization for worsening disease during the 6 weeks before enrollment; received and completed BNT162b2 COVID-19 vaccine series under EUA or approved dosing guidelines at least 6 months prior to study drug dosing.
[0676] Key inclusion criteria include known history of SARS-CoV-2 infection, prior administration of SARS-CoV vaccine other than the BNT162b2 COVID-19 vaccine series, orior administration of SARS-CoV-2 monoclonal antibody, history of severe adverse reaction associated with a vaccine or monoclonal antibody and/or severe allergic reaction (eg, anaphylaxis) to any component of the study intervention(s), bleeding diathesis of condition associated with prolonged bleeding that would m in the opinion of the investigator, contraindicate intramuscular injection.
[0677] Participants eligible for randomization are admitted to the clinical unit the morning of Day 1 and are confined in the clinical unit for a minimum of 1 hour following study drug administration. Each participant receives a single IM dose of the study drug.
[0678] Participants are required to complete an e-diary to monitor and record local reactions, systemic events and antipyretic medication usage for 7 days following administration of study drug. [0679] Additional safety assessments include monitoring of AEs, vital sign measurements, and physical examinations. Participants are monitored for all adverse events up to Day 90 and for SAEs and AESIs through 12 months via routine clinic visit, telehealth or telephone contact.
[0680] Serum virus binding and neutralizing antibody titers are assessed at specific timepoints through 12 months.
[0681] Participants are monitored for potential cases of COVID-19. If, at any time, a participant develops acute respiratory illness, the participant can contact the site and an in-person or telehealth visit should occur during which a SARS-CoV-2 RT-PCR test is conducted and serum virus neutralizing antibody titers are measured.
[0682] Blood samples are collected for PK and ADA throughout the follow-up period in ADI-58125 recipients only. Sampling timepoints for PK are determined based on modeling and simulation of human PK, using data generated with ADI-58125 in Phase 1 and Phase 2 studies.
[0683] Primary endpoints include:
• Geometric mean titer (GMT) of SARS-CoV-2 neutralizing activity on Days 7, 14, 28 and Months 2, 6 and 12
• Levels of SARS-CoV-2 spike protein binding antibodies, including antibodies to the NTD, RBD and S2 subunits on Days 7, 14, 28 and Months 2, 6 and 12
• Assessment of safety and tolerability of ADI-58125 based on the occurrence of treatment- emergent adverse events (TEAEs), local reactogenicity, systemic reactogenicity and use of antipyretics
• Changes from baseline in physical examination, vital signs (body temperature, pulse rate, respiration rate, and systolic and diastolic blood pressure)
[0684] Secondary endpoints include:
• Geometric mean-fold rise (GMFR) in neutralizing titer from baseline to 7, 14 and 28 days post dose
• Percentage of patients with > 4-fold rise in SARS-CoV-2 specific serum neutralizing titers on Days 7, 14 and 28
• Change from baseline in SARS-CoV-2 spike protein binding antibodies, including antibodies to the NTD, RBD and S2 subunits on Days 7, 14, 28 and Months 2, 6 and 12
• Incidence of RT-PCR confirmed, symptomatic COVID-19
• PK parameters of ADI-58125
• Immunogenicity of ADI-58125 (ADA response)
[0685] Exploratory endpoints include incidence of asymptomatic SARS-CoV-2 infection as determined by positive serology to N protein.
Immunology Analysis
[0686] For SARS-CoV-2 specific serum neutralizing titers, geometric mean titers and 2-sided 95% CIs are provided for each treatment group at baseline (prior to dosing) and on Days 7, 14, 28 and Months 3, 6 and 12.
[0687] Geometric mean titers (GMT) and the associated 2-sided CIs are derived by calculating means and CIs on the natural log scale based on the t-distribution, and then exponenting the results. [0688] GMFR and 2-sided 95% CIs are provided for each treatment group at the following timepoints: Days 7, 14 and 28; GMFRs are limited to participants with nomissing values at baseline and the specified post-dose time point. The GMFR is calculated as the mean of the difference of logarithmically transformed assay results (later time point - baseline) and transformed back to the original scale. Two-sided CIs are obtained by calculating CIs using Student’s t-distribution for the
mean difference of the logarithmically transformed assay results and transforming the limits back to the original scale.
[0689] Percentages (and 2-sided 95% CIs) of participants with > 4-fold rise are provided for each treatment group at the following timepoints: Days 7, 14 and 28. The Clopper-Pearson method is used to calculate CIs.
Safety Analysis
[0690] No formal statistical testing is performed on safety variables. Descriptive statistics for demographics and other baseline characteristics are provided. The Medical Dictionary for Regulatory Activities (MedDRA®) is used to classify all AEs with respect to system organ class and preferred term. Local and systemic reactogenicity, use of antipyretic medications and TEAEs are summarized for each cohort. The incidence of TEAEs, relationship to study drug, and severity are presented by system organ class and preferred term according to MedDRA coding dictionary. For vital signs, descriptive statistics are used to summarize for each cohort the observed results at each time point and the changes from baseline.
PK Analysis
[0691] Serum PK parameters are calculated for ADI-58125 using non-compartmental analysis methods. Descriptive statistics (min, max, median, mean, standard deviation, and coefficient of variation) of all PK parameters are provided.
[0692] Accordingly, this study provides evaluation of the magnitude of the SARS-CoV-2 neutralizing/binding antibody response in previously vaccinated adults receiving ADI-58125 supplementation compared to COVID-19 vaccine boost; the safety and tolerability of ADI-58125 supplementation; the pharmacokinetics and the immunogenecity of ADI-58125; and the incidence of RT-PCR confirmed, symptomatic COVID-19 in previously vaccinated adults receiving ADI-58125 supplementation compared to COVID-19 vaccine boost.
Example 11: Evaluation of ADI-58125 as a Preventive Option in Immunocompromised Individuals
[0693] This Example provides evaluation of ADI-58125 in an immunocompromised population in the prevention of symptomatic COVID-19.
[0694] This cohort includes significantly immune compromised adult participants with no known recent exposure but whose underlying medical condition(s) put them at increased risk of developing severe symptomatic COVID-19. Enrolled participants include, for example, subjects actively treated for solid tumor or hematologic malignancies, SOT recipient taking immunosuppressive therapy, CAR-T-cell therapy or HCT recipient, subjects having moderate or severe primary immunodeficiency, subjects having advanced or untreated HIV infection, or subjects currently taking high-dose corticosteroids, alkylating agents, antimetabolites, or TNF blockers.
[0695] Participants are assigned to receive a single IM dose of ADI-58125 (up to N=200).
The study evaluates the safety, tolerability, PK, and sVNA titers of ADI-58125 in significantly immune compromised adults in a single arm, open-label fashion.
Example 12: Evaluation of ADI-58125 as a Treatment or Preventive Option in Pediatric Population
[0696] This Examples provides evaluation of ADI-58125 in pediatric population. A separate open- label cohort including children from bith to 11 years of age is added to (1) the phase 2/3 randomized, double-blinded, placebo-controlled trial to evaluate the efficacy and safety of ADI-58125 in the treatment of ambulatory participants with mild or moderate COVID-19 (STAMP), and (2) the phase 2/3 randomized, double blind, placebo controlled trial to evaluate the efficacy and safety of ADI- 58125 in the prevention of COVID-19 (EVADE).
Treatment study
[0697] For the treatment study (STAMP), an open label treatment with ADI-58125 is given to children from birth to 11 years with mild or moderate COVID-19. The objective of this cohort is to evaluate the safety, tolerability and pharmacokinetics of ADI-58125 in children with mild to moderate COVID-19.
[0698] Three age groups, birth to <1 month, 1 month to <2 years and 2 years to 11 years are enrolled within this cohort .The proposed dose for study is expected to vary by age and will not exceed the anticipated adult treatment dose of 300 mg IM. For example, in one embodiment, the dosing regimen can be the below: a) 15 mg/kg for children from birth to < 1 month b) 10 mg/kg for 1 month to < 2 years c) 5 mg/kg for 2 to 11 years
[0699] Final dose selection is based on modeling and simulation, and is informed by the established pharmacokinetics of similar monoclonal antibodies and the pharmacokinetics of ADI-58125 observed in adults and adolescents. In addition, PK in children birth to 11 years of age is reviewed on an ongoing basis to ensure exposures are within the desired targets.
[0700] Children from birth to 11 years of age with symptoms of mild or moderate COVID-19 and a positive SARS-CoV-2 test are enrolled to receive a single IM dose of ADI-58125. Participants are enrolled simultaneously across age groups. ADI-58125 is administered at the study site and participants are monitored after dosing for acute worsening of disease, hypersensitivity, and injection site reactions. After Day 1, participants are monitored through Day 29 for safety, PK, viral titers and clinical outcome. In addition, participants continue long-term follow-up for safety, PK, ADA and development of post-acute sequelae of COVID-19 through approximately 5 half-lives.
Prevention study
[0701] For the prevention study (EVADE), an open label treatment with ADI-58125 is given to children from birth to 11 years at risk for developing SARS CoV-2 infections. The objective of this cohort is to evaluate the safety, tolerability and pharmacokinetics of ADI-58125 in children at risk for developing SARS CoV-2 infection.
[0702] Three age groups, birth to < 1 month, 1 month to <2 years and 2 years to 11 years will be enrolled. The proposed dose designed to achieve a minimum of 6-months protection in this study is expected to vary by age and will not exceed the anticipated adult treatment dose of 300 mg IM. For example, in one embodiment, the dose regimen can be the below: a) 10 mg/kg for birth to < 1 month (with a second 10 mg/kg dose 4 months post first dose) b) 10 mg/kg for 1 month to < 2yrs c) 5 mg/kg for 2 to 11 years
[0703] Final dose selection is based on modeling and simulation and is informed by the established pharmacokinetics of similar monoclonal antibodies and the pharmacokinetics of ADI-58125 observed in adults and adolescents. In addition, PK in children birth to 11 years of age is reviewed on an ongoing basis to ensure exposures are within the desired targets.
[0704] The preliminary sample size estimations for the planned pediatric studies are based on a predicted CV% of 30% in relevant PK parameters [e.g., area under the concentration-time curve (AUC) and maximum observed concentration (Cmax)]. It is expected that between 4 and 8 children for each age group will be enrolled to provide > 80% power to obtain reasonably precise estimates (i.e., 95% Cl with 60 and 140% of the geometric mean) of AUC and Cmax for each age group. To ensure adequate sample size in each age category, PK data for each age group are pooled across the two studies for analysis. It is anticipated that the total sample size includes 16 participants in the 2 to 11 and 1 month to < 2 year age groups and 8 participants in the birth to < 1 month age group.
[0705] Children from birth to 11 years of age are enrolled to receive a single IM dose of ADI-58125. Participants are enrolled simultaneously across age groups. ADI-58125 will be administered at the study site and participants will be monitored after dosing for acute hypersensitivity and injection site reactions. After Day 1, participants are monitored through approximately 5 half-lives for safety, PK, ADA and the development of SARS-CoV-2 infection (symptomatic or asymptomatic). Participants who develop signs or symptoms consistent with a COVID-like illness (CLI) are evaluated for laboratory confirmed COVID-19. Participants with laboratory confirmed COVID-19 are monitored for clinical and virological outcomes.
Example 13: Population Pharmacokinetics (PPK) of ADI-58125, an Extended-Half-life Monoclonal Antibody Being Developed for the Treatment and Prevention of COVID-19 [0706] This Examples provides a population pharmacokinetics (PPK) model that describes the serum ADI-58125 concentration-time profile following intravenous (IV) and intramuscular (IM) administration in Phase 1 and Phase 2/3 COVID-19 prevention and treatment studies.
[0707] The ADI-58125 PPK model was developed on PK data from a phase 1 first-in-human, single- ascending dose study (24 adults, IV and IM) and from phase 2/3 COVID-10 prevention (EVADE, 659 adults, IM) and treatment (STAMP, 189 adults, IM) studies. 1,486 PK samples were included in the analysis. Data from a phase 1 study were used to develop the base structural model describing the serum disposition of ADI-58125. The base structural model was adapted from a preliminary model developed using data from non-human primate studies. As feasible, the impact of body weight was included in the model, consistent with models for other mAbs. The model was qualified using visual predictive check (VPC) plots, and the precision of PPK parameters was evaluated using bootstrap/resampling methods. The impact of covariates (e.g. body weight, age, and baseline viral load) on ADI-58125 serum disposition were evaluated. Prediction-corrected visual predictive check (PC- VPC) plots were used to qualify the PPK model. Subject-specific estimates of ADI-58125 exposures were generated using individual, post hoc PK parameters.
[0708] The PPK model comprised 2 systemic compartments, zero-order infusion for IV administration and first-order absorption for IM administration, and provided a robust fit to the data based on the VPC plots (Figure 28) and goodness-of-fit plots (Figure 49). The impact of body weight on the variability in total body clearance, distributional clearance, and distributional volume parameters was evaluated and included in the model. A fitted exponent of 0.658 was found to be most appropriate for the relationship between body weight and the two clear ane terms, body clearance and distributional clearance, while a direct linear relationship with an exponent of 1.0 was most appropriate for the volume of the central and peripheral compartments.
[0709] Body weight influenced clearance, intercompartmental clearance, and central and peripheral volume compartments. The relationship between body weight and clearance was not suggestive of the need for dose adjustment over the population weight range studied (38.6 to 178.7 kg). There was no apparent impact of baseline viral load or age on ADI-58125 clearance. The median [range] half-lives in days by study; Phase 1 (a 1.71 [1.18-2.46]; b 125 [117-149]), Phase 2/3 prevention (a 1.86 [0.640- 3.13]; b 136 [105-209]), and Phase 2/3 treatment (a 1.89 [0.631-3.01]; b 136 [108-206]). The population mean IM bioavailability estimate was 90.5%. Figure 54 shows the PPK model median (90% confidence interval) concentration-time profile following a single 300 mg IM ADI-58125 dose by study.
[0710] The population pharmacokinetic model provides a precise and unbiased fit to observed ADI- 58125 concentration-time in healthy adults. The VPC plots shows that model-based simulations
recapture the observed data reliable (Figure 28). In addition, the traditional goodness-of-fit plots showed that the fit to the data is precise and unbiased both on a population mean and individual basis (Figure 49). The conditional weighted residual plots indicate that there is minimal bias relative to the population fitted concentrations and time since dose.
[0711] The post hoc empirical Bayesian PK parameter estimates were used to calculate derived PK parameters of AUC, Cmax and half-life, which are summarized in Table 16B below. The distribution of the dose-independent PK parameters in the 24 subjects included in the analysis are provided in
Figure 50.
AUC: area under the curve. Cmax: maximum serum concentration. CL: clearance. Tm· half-life. Vss: steady state volume of distribution.
[0712] The median population prediction of elimination half-life was 123 days, ranging from 79-277 days. The results are consistent with the intended PK characteristics of ADI-58125 (i.e., prolonged half-life and high IM bioavailability). The median AUC from time zero to 6 months and Cams increased in a dose-proportional manner when comparing the 300 mg and 600 mg IM groups. This is consistent with the linear elimination found to be most appropriate during population PK model development.
[0713] Figure 51 shows the population mean predicted concentrations overtime with IV or iM administration. At a dose of 300 mg, ADI-58125 had a robust IM bioavailability. Absorpotion from the IM depot resulted in lower peak concentrations than those seen with the IV depot, but profiles are similar after about 2 months. The population mean estimate of IM bioavailability was 92.2%, which is also confirmed by the population PK model.
[0714]
[0715] In conclusion, a 2-compartment PPK model with linear elimination and first-order IM absorption provided a precise and unbiased fit to the observed ADI-58125 concentration-time data. Moreover, ADI-58125 demonstrated high IM bioavailability and a median terminal elimination half- life of 125 to 136 days. The results are consistent with the intended PK characteristics of ADI-58125
in terms of prolonged half-life and high IM bioavailability. This PPK model is useful for future PK- pharmacodynamic analyses and simulations conducted to support phase 2/3 dose selection.
Example 14. Addressable Patient Populations for ADI-58125
Pre-exposure prophylasis
[0716] ADI-58125 can be used in adults and pediatric individuals:
• Who are not currently infected with SARS-CoV-2 and who have not had a known recent exposure to an individual infected with SARS-CoV-2 and o Who have moderate to severe immune compromise due to a medical condition or receipt of immunosuppressive medications or treatments and may not mount an adequate immune response to COVID-19 vaccination; or o For whom vaccination with any available COVID-19 vaccine, according to the approved or authorized schedule, is not recommended due to a history of severe adverse reaction (e.g., severe allergic reaction) to a COVID-19 vaccine(s) and/or COVID-19 vaccine component(s).
[0717] The following patient populations are also eligible to receiving ADI-58125:
• Patients who are within 1 year of receiving B-cell depleting therapies (e.g., rituximab, ocrelizumab, ofatumumab, alemtuzumab)
• Patients receiving Bruton tyrosine kinase inhibitors
• Chimeric antigen receptor T cell recipients
• Post-hematopoietic cell transplant recipients who have chronic graft versus host disease or who are taking immunosuppressive medications for another indication
• Patients with hematologic malignancies who are on active therapy
• Lung transplant recipients
• Patients who are within 1 year of receiving a solid-organ transplant (other than lung transplant)
• Solid-organ transplant recipients with recent treatment for acute rejection with T or B cell depleting agents
• Patients with severe combined immunodeficiencies
• Patients with untreated HIV who have a CD4 T lymphocyte cell count <50 cells/mm3
Treatment
[0718] The CDC COVID-19 Treatment Guidelines prioritize the following risk groups for anti- SARS-CoV-2 therapy based on 4 key elements: age, vaccination status, immune status, and clinical risk factors. The groups are listed by tier in descending order of priority, and these patient populations are eligible to receiving ADI-58125 as a treatment for COVID:
[0719] Tier 1:
• Immunocompromised individuals not expected to mount an adequate immune response to COVID-19 vaccination or SARS-CoV-2 infection due to their underlying conditions, regardless of vaccine status (see Immunocompromising Conditions below); or
• Unvaccinated individuals at the highest risk of severe disease (anyone aged >75 years or anyone aged >65 years with additional risk factors)
[0720] Tier 2:
• Unvaccinated individuals at risk of severe disease not included in Tier 1 (anyone aged >65 years or anyone aged <65 years with clinical risk factors)
[0721] Tier 3
• Vaccinated individuals at high risk of severe disease (anyone aged >75 years or anyone aged >65 years with clinical risk factors)
[0722] Tier 4
• Vaccinated individuals at risk of severe disease (anyone aged >65 years or anyone aged <65 with clinical risk factors).
Post-Exposure Prophylaxis
[0723] ADI-58125 may be used in adults and pediatric individuals for post-exposure prophylaxis of
COVID-19 in individuals who are at high risk for progression to severe COVID-19, including hospitalization or death, and are o not fully vaccinated or o who are not expected to mount an adequate immune response to complete SARS- CoV-2 vaccination (for example, individuals with immunocompromising conditions including those taking immunosuppressive medications) and
■ have been exposed to an individual infected with SARS-CoV-2 consistent with close contact criteria per Centers for Disease Control and Prevention (CDC) or
■ who are at high risk of exposure to an individual infected with SARS-CoV-2 because of occurrence of SARS-CoV-2 infection in other individuals in the same institutional setting (for example, nursing homes, prisons)
[0724] The following patient populations are also eligible to receiving ADI-58125 as a prophylactic alternative:
• Exposed patients concerned about “Long COVID” seeking to rapidly drive down their viral load in case they have COVID; and
• Exposed patients for whom quarantining is not an option due to impending travel, work obligations, or other reasons.
Pediatrics
[0725] Although children are at lower risk of developing severe COVID-19 compared to adults, a subset of children experience poor outcomes, including severe acute disease, such as the multisystem inflammatory syndrome, or MIS-C, and long-term sequelae of disease, also known as long COVID. Safe and effective therapies are needed to prevent severe disease and hospitalization in high-risk children as well as complications of COVID-19 such as MIS-C and long COVID. Similarly, although there is a paucity of data regarding the immune response to COVID-19 vaccines in children with moderate to severe immunocompromise, a subset of these children may have suboptimal immune responses to vaccines similar to adults with certain forms of immunocompromise and thus have the potential to benefit from a passive immune approach. Currently, the CDC recommends that children ages 5 to 11 with moderate to severe immunocompromise receive a 3 dose primary series of the Pfizer-BioNTech vaccine and that pre-teens and adolescents with moderate to severe immunocompromise receive a total of 4 doses of a mRNA COVID-19 vaccine. ADI-58125 can be used in pediatric individuals:
Example 15. Development of Broad and Potent Neutralizing Antibodies to Combat SARS-CoV- 2 and Future Emerging SARS-like Viruses
[0726] The neutralizing antibodiy response to SARS-CoV-2 is dominated by three classes of public RBD antibodies which share common escape mutations (Figures 29A and 29B). A newly emerged Omicron (B.1.1.529/BA.1) variant contains mutations in the epitopes of all three public classes of antibodies (Figure 30). Conserved spike mutations in the Omicron (B.1.1.529/BA.1) variant include A67V, D69-70, T95I, G142D/A143-145, A211/L212I, ins214EPE, G339D, S371L, S373P, S375F, K417N, N440K, G446S, S477N, T478K, E484A, Q493K, G496S, Q498R, N501Y, Y505H, T547K, D614G, H655Y, N679K, P681H, N764K, D796Y, N856K, Q954H, N969K, and L981F (Figure 30). [0727] Many clinical stage/EUA authorized antibodies target common epitopes, such as LY-C0VOI6 (etesevimab), REGN10933 (casirvimab) (both in class 1); LY-CoV555 (bamlanivimab), ADZ8895 (COV2-2196 or tixagevimab), CT-P59 (regdanvimab) (ah in class 2); and REGN10987 (imdevimab) and AZD1061 (COV2-2130 or cilgavimab) (both in class 3). Escape mutations from these antibodies are determined by deep mutational scanning as described previously; in particular, E484 and Q493 for Ly-Cov555; K417 for Ly-CoV16; K417, E484, and Q493 for REGN10933; G446 and N440 for REGN10987; G446 for AZD1061; and G339 for sotrovimab, among others (Starr T., et al, Science, 2021; Greany, A. et al., Nat. Comm., 2021; Dong J. et al, Nat. Micro., 2021). As shown in Figures 30, 31, 32A and 32B, ADI-58125 binds to an epitope distinct from those recognized by commonly elicited nAbs. The inherent features of the claimed broadly neutralizing antibodies, such as ADI-
58125, offer a high barrier to resistance by targeting of immunoquiescent epitopes (protected from immune pressure) and recognition of highly conserved residues (likely important for viral fitness). [0728] Figure 41 depicts the average fold change in IC50 relative to the D614G reference strain of ADI-58125 in a lenti viral pseudo virus assay. All spike variants contain the D614G substitution. To minimize the impact of potential assay variability, D614G was tested in parallel with each variant, and the variant-matched D614G IC50 value was used to calculate the fold change in IC50. Variants of concern and their descendent lineages are highlighted in red. Data represents the mean of at least two indepdendent experiments, and error bars represent standard deviation. As shown in Figure 41, ADI- 58125 exhibited a 328-fold loss in neutralization activity against the newly emergent B.1.1.529/BA.l/Omicron variant of concern. Similarly, B.1.1 -529/Omicron pseudovirus containing the R346K mutation, which is currently present in approximately 10% of sequenced Omicron variants, led to a 286-fold reduction in IC50 relative to D614G. However, despite the reduction in neutralization activity, ADI-58125 still maintained neutralization potency against Omicron. Figure 42A_shows the neutralization curves for ADI-58125 and ADI-58122 against Omicron (BA.l) and D614G (also referred to as wild type, WT), and Figure 42B_shows the neutralization curves for ADI- 58125 against D614G and Omicron sublineages (BA.l, BA.1.1, and BA.2) using a pseudovirus neutralization assay. As shown in Figure 42A, ADI-58125 retained partial potency against theOmicron (BA.l) variant, with an IC50 against Omicron of about 211.8 ng/mL and an IC50 against D614G of about 0.6 ng/ml. The IC50 for ADI-58122 was shown to have an IC50 of about 8.5 ng/mL for D614 and about >4000 ng/ml for the Omicron variant. Figure 42B also shows the in vitro neutralizing activity of ADI-58125 against the Omicron BA.l and BA.1.1 sub-lineages.
[0729] Further in vitro studies examined the neutralization potencies of large panels of mAbs against the Omicron variant (B.1.1.529/BA.1) in both authentic and pseudovirus assays. Findings across the studies show that among mAbs in late-stage clinical development or with Emergency Use Authorization (EUA), ADI-58125 is one of only a few mAbs that demonstrated neutralizing activity against Omicron. Across two distinct authentic neutralization assays against Omicron, the data show that ADI-58125 had an IC50, a measurement of neutralization potency, of approximately 0.2 to 1.1 μg/mL and an IC90 of about 9.943 μg/mL. See also, e.g., Dejnirattisai et al. (2022) SARS-CoV-2 Omicron-B.1.1.529 leads to widespread escape from neutralizing antibody responses; Planas et al. (2021) Considerable escape of SARS-CoV-2 variant Omicron to antibody neutralization; Liu et al. (2021) Strikig antibody evasion manifested by the Omicron variant of SARS-CoV-2; Cameroni et al. (2021) Broadly neutralizing antibodies overcome SARS-CoV-2 Omicron antigenic shift; and Lusvarghi et al. (2022) Comparison of SARS-CoV-2 Omicron neutralization by therapeutic antibodies, convalescent sera, and post-mRNA vaccine booster sera, the entire contents of each of which are expressly incorporated herein by reference.
[0730] Additional binding and neutralization assays were performed on Victoria and all variants of concern, Alpha, Beta, Gamma, Delta and Omicron (BA.l., BA.1.1, and BA.2). Omicron (where not specified Omicron refers to sub-lineage BA.l) contains an unprecedented number of mutations concentrated in the Spike (S) gene which carries 30 substitutions plus the deletion of 6 and insertion of 3 residues. BA.1.1 has an additional R346K mutation than BA.l. BA.2 differs from BA.l at 6 positions in the RBD. A slightly increased affinity of BA.2 RBD for ACE2 was observed, which may underlie its increased transmissibility. Neutralization of BA.l to BA 1.1 and BA.2 was also compared, where both BA.1.1 and BA.2 were shown to be modestly more difficult to neutralize than BA.l using vaccine serum.
[0731] The neutralization potential of antibodies was measured using a Focus Reduction Neutralization Test (FRNT). Briefly, the reduction in the number of the infected foci is compared to a negative control well without antibody. Briefly, serially diluted Ab or plasma was mixed with SARS- CoV-2 strains and incubated for 1 hr at 37°C. The mixtures were then transferred to 96-well, cell culture -treated, flat-bottom microplates containing confluent Vero cell monolayers in duplicate and incubated for a further 2 hrs followed by the addition of 1.5% semi-solid carboxymethyl cellulose (CMC) overlay medium to each well to limit virus diffusion. A focus forming assay was then performed by staining Vero cells with human anti-NP mAh (mAb206) followed by peroxidase- conjugated goat anti-human IgG (A0170; Sigma). Finally, the foci (infected cells) approximately 100 per well in the absence of antibodies, were visualized by adding TrueBlue Peroxidase Substrate. Virus-infected cell foci were counted on the classic AID EliSpot reader using AID EFISpot software. The percentage of focus reduction was calculated and IC50 was determined using the probit program from the SPSS package.
[0732] Authentic neutralization assays were performed for antibodies being developed for commercial use (Figures 43 and 44). As shown in Figures 43 and 44, although neutralization of BA.2 was knocked out for ADI-58125, it was still able to neutrali e BA.l and BA.1.1. See also, e.g., Nutalai et al, (2022) Potent cross-reactive antibodies following Omicron breakthrough in vaccinees, the entire contents of which are expressly incorporated herein by reference.
[0733] These data uniformly support that ADI-58125 is among the few mAbs to demonstrate neutralizing activity against the Omicron variant, and also consistently support the findings from a variety of preclinical studies showing that ADI-58125 retains activity against other variants of concern including Alpha, Beta, Delta and Gamma, and that ADI-58125 retains neutralizing activity against a diverse panel of circulating SARS-CoV-2 variants, including the Fambda, Mu and Delta plus variants, thus, warranting its continued development.
Sarbecovirus cross-reactivity binding assay
[0734] ADI-58125 and human ACE2 (hACE2; expressed in a bivalent format as a C-terminal IgGl Fc conjugate with an approximate molecular weight of 290 kDa; Sino Biological, Cat # 10108-H02H)
binding to surface-displayed RBD was first assessed at a single 100 nM concentration against the panel of 17 yeast-surface displayed sarbecovirus RBD-SD1 proteins. 0.1 OD induced cells per well were aliquoted into 96-well plates and washed out of induction media with PBSF (PBS with 0.1% w/v BSA). Next, cells were resuspended in 100 pL of ADI-58125 at 15 μg/mL or hACE2 at 29 ug/ml and incubated at 24 °C for 30 minutes, followed by two washes with PBSF. Cells were subsequently labeled with 50 pL of 1:100 diluted allophycocyanin (APC)-conjugated monoclonal mouse anti hemagglutinin tag (HA).11 antibody (BioLegend, Cat # 901524), 1:100 diluted phycoerythrin (PE)- conjugated goat anti-human IgG polyclonal antibodies (Southern Biotech, Cat # 2040-09), and 5 μg/mL propidium iodide (Invitrogen, Cat # P1304MP); and incubated on ice for 20 minutes. For each sarbecovirus, an additional sample was labeled with only secondary reagents to establish background signal levels. Cells were washed out of secondary staining reagents with PBSF twice before analyzing via flow cytometry on a BD FACS Canto II. Normal and live yeast cells (as determined by forward scatter-side scatter (FSCxSSC) and propidium iodide incorporation, respectively) were collected for each sample to measure mean fluorescence intensity (MFI) of surface RBD-SD1 expression and ADI- 58125/ACE2 binding.
SARS-CoV-2 variants binding assay
[0735] SARS-CoV-2 variants commonly present in GISAID were evaluated for ADI-58125/ADI- 58122 binding. To more comprehensively assess the impact of different substitutions at D405, G502, G504, and Y505 residues, RBDs incorporating ah 19 substitutions at each residue were generated. Antibody binding experiments were performed essentially as described with sarbecovirus binding, with the difference that yeast cells were stained with ADI-58125 at its KDApp concentration, 0.43 nM, to maximize the dynamic range of differences in binding affinity to variants compared to the Wuhan-Hu-1 RBD-SD1. Binding signal was normalized N-terminal HA expression signal and background staining by dividing mean PE MFI by the mean APC MFI and subtracting the PE/APC ratio of non-primary labeled cells and expressed as a percentage of SARS-CoV-2 wildtype binding by ADI-58125.
[0736] Affinity maturation of ADI-58125 against the SARS-CoV-2 BA.l Omicron variant was performed by introducing diversities into the heavy chain and light chain variable regions. Briefly,
VH and VF CDR diversification was obtained by odering forward priming oligos with variation in each cdr. FR1-FR4 oligos containing homology to the CDRs above were ordered in the reverse priming direction for the assembly and amplification of the entire heavy or light chain variable regions via PCR. The heavy chain variable regions (FR1-FR4) were transformed into yeast containing the light chain plasmid of the parent. The light chain variable regions (FR1-FR4) were transformed into yeast containing the heavy chain plasmid of the parent. Selections were performed with rounds of FACS, sorting for the highest affinity biotinylated SARS-CoV-2 spike protein binders using antigen titration.
[0737] Figure 52A provides a graph comparing binding affinity of ADI-58125 and affinity maturation progenies for the SI from the Omicron BA.l varaint. Figure 52B provides a graph comparing the ability of ADI-58125 and affinity maturation progenies to neutralize the Omicron BA.l varaint. As shown in Figure 52A, affinity maturation progenies were able to increase the binding affinity against the Omicron BA.l SI protein by over 100-fold (KD of 291 nM for ADI-58125 vs. KD of 2.11 nM for affinity maturation progenies). Similarly, affinity maturation progenies were able to improve the neutralization against the Omicron BA.l varaint by about 40 folds (ICso of 0.215 μg/mL for ADI-58125 vs. IC50 of 0.0.005 μg/mL for affinity maturation progenies), while maintaining activity against other VOCs and SARS-CoV (Figure 52B).
[0738] Figure 53A provides a graph comparing binding affinity of re-engineered ADI-58125 progenies for various SARS-CoV-2 variants, including the BA.l Omicron variant, the Beta variant, the Delta variant and D614G, and SARS-CoV. Figure 53B shows the corresponding neutralization IC50 of re-engineered ADI-58125 progenies. As shown in Figures 53A and 53B, affinity matured ADI-58125 progenies maintained binding and neutralizing activities against the variants of concern.
Example 16. Efficacy of ADI-58125 Against SARS-CoV-2 in Rhesus Macaque Intratrachel- intranasal Exposure Model
[0739] This study demonstrated ADI-58125 treatment offers protection against SARS-CoV-2 infection, resulting in only minimal to mild pulmonary inflammation. Rhesus macaque were exposed to SARS-CoV-2 after receiving ADI-58125 (25 mg/kg or 5 mg/kg), or irrelevant control monoclonal antibody (mAh) (5 mg/kg), 3-days prior. Evaluation of microscopic pulmonary lesions among the groups with comparing and contrasting the findings aims to support the development of an mAh that provides prophylactic and therapeutic benefit.
[0740] Methods and Materials
[0741] A total of 12 rhesus macaques were included in the study. The study was comprised of 3 groups: group 1 received 25 mg/kg Control mAh; group 2 received 25 mg/kg ADI-58125; and group 3 received 5 mg/kg ADI-58125 Each group had 4 NHPs of the same species that were randomly assigned, balanced by sex and body weight. See Table 17.
[0742] Challenge agent: Groups 1-3 SARS-CoV-2
[0743] Target Challenge Dose/Route: IT/IN exposure 1 x 106 pfu.
[0744] All necropsies were performed by a veterinary pathologist within 5 hours of euthanasia, study days 5 and 6 respectively. All necropsies were performed IAW Pathology Division SOPs. All gross findings were recorded as appropriate in the currently validated version of Pristima. Tissues were collected IAW Pathology Division SOPs and fixed by immersion into labeled containers of 10% neutral buffered formalin for the specified time according to the currently validated pathogen-specific
inactivation time. Tissues were collected for each animal. Additional tissues were also collected when gross observations warrant histopathologic evaluation.
[0745] Tissues were removed from biocontainment IAW Pathology Division SOPs and transported to the USAMRIID histology lab for processing. The tissues were trimmed, processed, embedded in paraffin, cut by microtomy, stained, coverslipped and screened IAW Pathology Division SOPs. Tissue blocks and slides were produced.
[0746] Replicate lung, nasal turbinates, trachea, larynx, tracheobronchial lymph node and olfactory bulb tissue sections were placed on positively-charged slides and stained by immunohistochemistry (IHC) procedures IAW Pathology Division SOPs.
[0747] All slides were examined by the veterinary pathologist and all findings were recorded into the currently validated version of Pristima. Following the conclusion of slide evaluation, the specimens were stored or archived by USAMRIID personnel IAW Pathology Division SOPs.
[0748] Results
[0749] All animals survived to study day 5 or 6 and were euthanized at the end of the study. Animals surviving to and euthanized at the end of the study were referred to as survivors. Survivor data was detailed in Table 17. Table 17. Animal Table
[0750] Necropsy Findings
[0751] Group 1: The most consistent gross finding in this group was enlarged (1.5 times to 3 times normal) tracheobronchial lymph nodes (present in 3/4 NHPs in the group). Additional gross findings in this group that at the time of necropsy were suspected to be disease or challenge related included moderate red discoloration and consolidation of the hilar region of the right caudal lung lobe (present 1/4 NHP (1221) in this group) (see Figures 33 and 34). Mild multifocal red discoloration was noted in all lung lobes (present 1/4 NHP (1294) in this group) (see Figure 39). Mild multifocal red discoloration was noted in the lower left nasal turbinate (present 1/4 NHP (624R) in this group). [0752] Group 2: The most consistent gross finding in this group was enlarged (1.5 times to 3 times normal) tracheobronchial lymph nodes (present in 3/4 NHPs in the group). Additional gross findings in this group that at the time of necropsy were suspected to be disease or challenge related included mild multifocal red discoloration noted in all lung lobes (present 1/4 NHP (0246) in this group) and in the left cranial and caudal lung lobes (present 1/4 NHP (8011) in this group).
[0753] Group 3: The most consistent gross finding in this group was enlarged (1.5 times to 3 times normal) tracheobronchial lymph nodes (present in all NHPs in the group). Additional gross finding in this group that at the time of necropsy were suspected to be disease or challenge related included mild multifocal red discoloration noted in all lung lobes along with a locally extensive fibrinous adhesion from the left caudal lung lobe to the thoracic wall (present 1/4 NHP (5240) in this group) (see Figure 40).
[0754] Other recorded gross necropsy findings in these groups are considered not to be disease or challenge related.
[0755] Microscopic Findings
[0756] A summary of the major histopathologic findings in the lungs graded by severity for each NHP was present in Table 18. A summary of the histological findings in the nasal turbinates graded by severity for each NHP was present in Table 19.
[0757] Group 1: The disease-related microscopic lesions present in this group were noted predominately in the lung. Additionally, nasal turbinates and the trachea had lesions that were suspected to be either challenge or disease related. All NHPs in this group had mild to moderate multifocal interstitial (expansion of the alveolar septae), perivascular and/or peribronchiolar pulmonary inflammation composed predominately of lymphocytes and histiocytes with fewer neutrophils. Additional pulmonary findings present in 3/4 NHPs in this group were minimal to mild type 2 pneumocyte hyperplasia and 2/4 NHPs (1221 and 642R) had mild fibrin deposition either within alveolar lumina or expanding the alveolar septa. IHC was multifocally minimally to mildly positive in areas of inflammation in all NHPs in this group (see Figure 35).
[0758] All NHPs in this group had mild to moderate inflammation in one or more sections of the nasal turbinates composed predominately of lymphocytes and plasma cells with fewer neutrophils with areas of erosion or ulceration, degeneration and necrosis and edema (see Figure 36). All NHPs in this group had minimal to mild inflammation in the submucosa of the trachea composed of lymphocytes, plasma cells and neutrophils. Rare epithelial cells lining the trachea were IHC positive in 2 NHPs (7137 and 1221) in this group along with rare epithelial cells lining the nasal turbinates were IHC positive in 1 NHP (7137). IHC as negative in all of the sections of nasal turbinates for 3/4 NHPs in this group.
[0759] All NHPs in this group had mild multifocal sinus histiocytosis in the tracheobronchial lymph node. Two NHPs in this group (642R and 1221) were also draining neutrophilic inflammation. IHC was negative in all sections of the tracheobronchial lymph node and in one olfactory bulb (1221) examined in all NHPs in this group. Three olfactory bulbs (642R, 1294 and 7137) and all larynx tissue were missing at time of trimming.
[0760] Group 2: The suspected disease -related microscopic lesions in this group were noted predominately in the lung. Additionally, nasal turbinates and the trachea had lesions that were suspected to be challenge related. The principle histological lesion present in the lungs of all animals in this group was multifocal and/or focal minimal to mild inflammation composed predominately of histiocytes and lymphocytes with fewer neutrophils. Of the eight sections of lung lobes examined in each NHP the multifocal mild inflammation was present in two sections (0246) and in one section (7175 and 6334). Multifocal minimal inflammation was present in two sections (6334) and in one section (8011). Focal minimal inflammation was also noted in one section of the eight examined (6334). The inflammation was present around vessels and it expanded alveolar septa and occasionally extended into the alveolar lumina in all animals. IHC was negative in all areas of inflammation in the lung tissue examined in all NHPs in this group (see Figure 37).
[0761] All NHPs in this group had mild to marked inflammation in one or more sections of the nasal turbinates composed predominately of lymphocytes and plasma cells with fewer neutrophils with areas of erosion or ulceration, degeneration and necrosis and edema. Three out of four NHPs (7175, 6334 and 8011) in this group had minimal submucosal inflammation in the trachea composed of lymphocytes, plasma cells and neutrophils. One NHP in this group (6334) had multifocal mild inflammation in the submucosa of the larynx composed of lymphocytes, plasma cells and neutrophils. IHC was negative in all of the sections of nasal turbinates, trachea and larynx.
[0762] All NHPs in this group had mild multifocal sinus histiocytosis in the tracheobronchial lymph node. IHC was negative in all sections of tracheobronchial lymph node, olfactory bulbs (8011, 7175, 6334) and larynx (8011, 7175, 6334). Olfactory bulb and larynx was missing at time of trim for NHP 0246.
[0763] Group 3: The suspected disease-related microscopic lesions in this group were noted predominately in the lung. Additionally, nasal turbinates and the trachea had lesions that were suspected to be challenge related. The principle histological lesion present in the lungs of 3/4 animals in this group was multifocal and/or focal minimal to mild inflammation composed predominately of histiocytes and lymphocytes with fewer neutrophils. Of the eight sections of lung lobes examined in each NHP the multifocal mild inflammation was present in one of the eight sections examined (5240, 1430 and 3471). Multifocal minimal inflammation was present in one sections (3471). The inflammation was present around bronchi or bronchioles (3471 and 5420), and it expanded alveolar septa and occasionally extended into the alveolar lumina (3471 and 1430). IHC was negative in all areas of inflammation in all NHPs in this group (see Figure 38).
[0764] All NHPs in this group had mild to moderate inflammation in one or more sections of the nasal turbinates composed predominately of lymphocytes and plasma cells with fewer neutrophils with areas of erosion or ulceration, degeneration and necrosis and edema. All NHPs in this group had minimal to mild inflammation in the submucosa of the trachea composed of lymphocytes, plasma cells and neutrophils. One NHP (3471) had a focal erosion of the tracheal mucosa. Three out of the four NHPs in this group (8075, 5240 and 1430) had multifocal mild inflammation within the submucosa of the larynx. IHC was negative in all of the sections of nasal turbinates, trachea and larynx examined for all NHPs in this group.
[0765] Two NHPs in this group had minimal to mild lymphoid hyperplasia and all NHPs had mild multifocal sinus histiocytosis in the tracheobronchial lymph node. IHC was negative tracheobronchial lymph node and olfactory bulb for all NHPs in this group. Olfactory bulb and larynx was missing at time of trim for NHP 3471.
[0766] Other recorded microscopic findings occurred in these groups and are considered incidental findings and are not disease related.
[0767] Summary
[0768] Gross to Microscopic Correlations
[0769] Group 1: The moderate red discoloration and consolidation of the hilar region of the right caudal lung lobe in NHP 1221 did correlate to disease related microscopic findings (see Figure 33). Extensive area of inflammation, edema, type II pneumocyte hyperplasia and fibrin deposition were noted within peripheral and central sections taken from the right caudal lung lobe. These lesions were IHC positive. The multifocal red discoloration noted throughout the lung lobes in NHP 1294 may correlate to disease related microscopic findings. In the affected lobes there was hemorrhage, edema and minimal to mild inflammation (all central sections examined and one peripheral sections examined). Lung tissue from this NHP was IHC positive. The grossly enlarged tracheobronchial lymph nodes in 3/4 NHPs (1294, 642R and 1221) in this group correlated to mild sinus histiocytosis this finding was most consistent with a reactive lymph node. Reactive lymph nodes had increased lymphatic flow which was evident by increased drainage from tissue by the presence of increased numbers of phagocytic cells (macrophages/histiocytes) within the medullary sinuses (sinus histiocytosis). This microscopic finding was likely evidence of antigenic stimulation from SARS- CoV-2 viral challenge. The red discoloration present on the lower left turbinate of NHP 642R did not correlate to any disease related microscopic findings.
[0770] Group 2: The multifocal mild red discoloration noted in all lung lobes (0246) and limited to the left cranial and caudal lung lobes (8011) did not correlate to disease related microscopic findings. Microscopically these sections of lung had either edema or congestion which were considered non specific histological findings. Grossly enlarged tracheobronchial lymph nodes in 3/4 NHPs (0246, 7175 and 8011) in this group correlated to mild sinus histiocytosis this finding was most consistent with a reactive lymph node. Reactive lymph nodes had increased lymphatic flow which was evident by increased drainage from tissue by the presence of increased numbers of phagocytic cells
(macrophages/histiocytes) within the medullary sinuses (sinus histiocytosis). This microscopic finding was likely evidence of antigenic stimulation from SARS-CoV-2 viral challenge.
[0771] Group 3: The pulmonary adhesions present in NHP 5240 from the left caudal lung lobe to the left thoracic wall and multiple areas of red discoloration did not correlate to disease related microscopic findings. Fibrous adhesions were noted between lung lobes microscopically; however, there were no microscopic changes to the underlying pulmonary parenchyma in the areas of pleural fibrosis and IHC was negative. Pleural or subpleural fibrosis with or without lung adhesions to adjacent lung lobes or the thoracic cavity were commonly encountered in the lungs of macaques. Grossly enlarged tracheobronchial lymph nodes in all NHPs in this group correlated to minimal to mild lymphoid hyperplasia in two NHPs (8075 and 5240) and mild sinus histiocytosis in all NHPs. These histological findings were most consistent with a reactive lymph node. Reactive lymph nodes had increased lymphatic flow which was evident by increased drainage from tissue by the presence of increased numbers of phagocytic cells (macrophages/histiocytes) within the medullary sinuses (sinus histiocytosis). This microscopic finding was likely evidence of antigenic stimulation from SARS- CoV-2 viral challenge.
[0772] Histopathology
[0773] Group 1: All NHPs in this group had mild to moderate lymphoplasmacytic and neutrophilic inflammation in one or more sections of the nasal turbinates. 2/4 NHPs (7137 and 642R) had mild submucosal edema, 2 NHPs had ulceration and/or erosion of the nasal epithelium (7137 and 1294) and two NHPs had degeneration and necrosis within the mucosa or submucosa (7137 and 1294). The cause of the inflammation and other microscopic findings were not evident histologically. It was suspected that these findings were the result of SARS-CoV-2 infection within the nasal turbinates as noted by the presence of few epithelial cells being IHC positive in NHP 7137. However, the inflammation could also be the related to the manipulations associated with experimental challenge route.
[0774] The other microscopic finding in this group that was suspected to be related to either the result of SARS-CoV-2 infection or experimental challenge route/manual manipulations (broncho- alveolar lavage performed on days 1 and 3 of the study) was the multifocal to mild inflammation noted in the trachea. IHC as positive in occasional tracheal epithelium in 2/4 NHPs (1221 and 7137) in this group.
[0775] Group 2: All NHPs in this group had mild to marked lymphoplasmacytic and neutrophilic inflammation in one or more sections of the nasal turbinates. The cause of the inflammation was not evident histologically. It was suspected that these findings were the result of SARS-CoV-2 infection within the nasal turbinates. However, the inflammation could also be the related to the manipulations associated with experimental challenge route.
[0776] Other microscopic finding in this group that were suspected to be related to experimental challenge route/manual manipulations (broncho-alveolar lavage performed on days 1 and 3 of the study) were present in the trachea and larynx. Three out of four NHPs (7175, 6334 and 8011) in this group had minimal submucosal inflammation in the trachea. One NHP in this group (6334) had multifocal mild inflammation in the submucosa of the larynx. IHC was negative in all sections of trachea and larynx examined.
[0777] Group 3: All NHPs in this group had mild to moderate lymphoplasmacytic and neutrophilic inflammation in one or more sections of the nasal turbinates. The cause of the inflammation or focal ulceration was not evident histologically. It was suspected that these findings were the result of SARS-CoV-2 infection within the nasal turbinates. However, the inflammation could also be the related to the manipulations associated with experimental challenge route.
[0778] Other microscopic finding in this group that were suspected to be related to experimental challenge route/manual manipulations (broncho-alveolar lavage performed on days 1 and 3 of the study) were present in the trachea and larynx. All NHPs in this group had minimal to mild inflammation in the submucosa of the trachea with a focal area of mucosal erosion noted in one NHP (3471). Three out of the four NHPs in this group (8075, 5240 and 1430) had multifocal mild inflammation within the submucosa of the larynx.
[0779] Conclusion
[0780] The gross findings in the lungs in one NHP in group 1 (1221), histological findings present in the lungs in 3/4 NHPs in group 1 and IHC results in all animals in group 1 parallel reported histopathological findings in rhesus macaques with SARS-CoV-2 infection (Munster VJ, et al.
Nature. 2020 Sep;585(7824):268-72). All animals were affected with varying degrees of pulmonary inflammation on 2 to 5 of the 8 sections of lungs examined. Positive immunohistochemistry present in all animals in this group in the areas of inflammation and within the respiratory epithelium supported SARS-CoV-2 as the underlying cause for the microscopic findings in the lungs in this group.
[0781] The predominate microscopic finding in the lungs of all group 2 NHPs was multifocal minimal to mild inflammation on 1 to 3 of the 8 sections of lungs examined. No other significant microscopic lesions were present in the lungs. Additionally IHC was negative on all sections of lungs examined for all NHPs in this group. It was suspected that the antibody treatment offered some protection resulting in only minimal to mild pulmonary inflammation on the histological section examined for this group.
[0782] The predominate microscopic finding in the lungs of 3/4 group 3 NHPs was multifocal minimal to mild inflammation on 1 to 2 of the 8 sections of lungs examined. No other significant microscopic lesions were present in the lungs. It is important to note that one NHP (8075) did not have the pulmonary inflammation typically associated with SARS-CoV-2 infection. Additionally IHC
was negative on all sections of lungs examined for all NHPs in this group. It was suspected that the antibody treatment offered some protection resulting in only minimal to mild pulmonary inflammation on the histological section examined for this group.
Example 17. Omicron-Based Dose Selection of ADI-58125 for the Prevention and Treatment of SARS-CoV-2
[0783] In the primary analysis population, the pre-Omicron group, patients infected with or exposed to a non-Omicron variant, ADI-58125 met the primary objectives with statistical significant across all three indications (pre- and post-exposure prophylaxis and treatment of COVID-19), demonstrating statistically significant and clinically meaningful efficacy. In pre -exposure and post-exposure prophylaxis, ADI-58125 was associated with 71% and 75% relative risk reductions compared to placebo, respectively, in the prevention of RT-PCR confirmed symptomatic COVID-19. In an exploratory analysis of the post-Omicron group, patients exposed to the Omicron variant, in pre exposure prophylaxis, ADI-58125 was associated with a clinically meaningful reduction in the risk of developing RT-PCR confirmed symptomatic COVID-19 compared with placebo. In treatment, ADI- 58125 was associated with a 66% relative risk reduction compared to placebo in the incidence COVID-19 related hospitalization or all cause death through Day 29. In patients treated within three days of symptom onset, ADI-58125 was associated with a reduced risk of COVID-19 hospitalization or death from any cause through Day 29 by 77% compared to placebo. A preliminary analysis of available safety data in each trial revealed no safety concerns for ADI-58125.
Prevention of SARS-CoV-2: ADI-58125-PREV-001 (EVADE)
[0784] In order to determine an appropriate pharmacodynamic target for prevention, available clinical data from vaccine and other monoclonal antibody (mAh) programs were evaluated to determine a neutralizing antibody titer threshold associated with protection against symptomatic COVID-19. To perform this analysis, published vaccine efficacy data (Polack 2020; Abu-Raddad 2021; Andrews 2021; AstraZeneca 2021; Clemens 2021; Emary 2021; Falsey 2021; Hitchings 2021; Liu 2021a; Lopez Bernal 2021; Madhi 2021; Nasreen 2021; O'Brien 2021; Tartof 2021; UKHSA
2021) were utilized, as well as geometric mean serum neutralization IC50 values for samples obtained fromChAdOxl nCoV-19/AZD1222 (AstraZeneca) and BNT162b2 (BioNTech/Pfizer) vaccinees at different time points following administration and against different variants of concern (AstraZeneca 2021; Liu 2021a). All of the vaccinee serum samples were evaluated using the same authentic virus neutralization assay. This assay was also used to determine the neutralization IC50 values for ADI- 58125, other clinical-stage mAbs, and mAbs authorized for emergency use (including REGEN -COV [Regeneron] and AZD7442 [AstraZeneca]) against an ancestral strain of SARS-CoV-2 (Victoria) as well as all variants of concern (Alpha, Beta, Gamma, Delta, and Omicron) (Liu 2021a; Dejnirattisai
2022). This allowed for a direct comparison of measured vaccine-induced neutralizing antibody titers
and projected serum neutralizing titers following passive antibody administration, which was then correlated to the respective reported efficacies (AstraZeneca 2021; O'Brien 2021; Regeneron 2021). [0785] The projected serum neutralizing titers for the mAbs were calculated by dividing the neutralization IC50 values by the estimated serum antibody concentration at a given timepoint following antibody administration based on internal (ADI-58125) or published (REGEN -COV and AZD7442) pharmacokinetic data (FDA 2020; Loo 2021). Based on the correlation between serum neutralizing antibody titers and reported vaccine and mAh efficacy against symptomatic COVID-19 (Figure 45), a threshold serum neutralizing titer of -1:100 was associated with -70% protection against symptomatic disease relative to placebo. Given the in vitro IC50 value of 1.029 μg/mL for ADI-58125 against the Omicron variant in this assay, a serum ADI-58125 concentration of - 100 μg/mL would be required to reach this threshold neutralizing titer.
[0786] To evaluate the dosing regimens of ADI-58125, a preliminary population PK (PopPK) model was developed based on data from a phase 1, first-in-human (FIH), single-ascending dose study of ADI-58125 in 24 healthy adult participants. The PopPK model utilized a two-compartment model with linear clearance (CL) from the central compartment. For IM administration, a first-order rate constant was used to describe the extravascular absorption. CL and volume of distributions were ahometricahy scaled from parameters. The median and 80% (10-90%) prediction interval (PI) were estimated from 500 randomly simulated subjects based on weight distribution of the subjects enrolled in the pivotal trial (EVADE or STAMP).
[0787] The 1200 mg IM dose for evaluation of the prevention of symptomatic COVID-19 in EVADE was chosen based on the projected ability to achieve the following targets after the initial absorption phase: 1) a median concentration of ADI-58125 (100 mg/L) in serum for three months (or 12 weeks) that is projected to be associated with a serum virus neutralizing titer of -1:100 as estimated above based on IC50 value; and 2) a median concentration (99.4 mg/L) of ADI-58125 in serum for three months (or 12 weeks) after dosing that is projected to achieve 90% neutralizing concentration (IC90, 9.94 μg/mL) against the Omicron variant in the epithelial lining fluid (ELF), assuming an ELF:serum ratio of -10% (Wohacott 2016; Deng 2018) (Figure 46). The IC50 and IC90 values generated against authentic virus were used for dose selection to maintain consistency across prior dose selection efforts.
[0788] As shown in Figure 45, predicted month 3 serum neutralizing antibody titers against the Omicron variant following IM administration of 1200 mg ADI-58125 exceed the neutralization titers predicted for AZD7442 following the single 300 mg IM dose currently authorized under EUA. In order to provide protection beyond the initial 3 months, a re-dosing strategy for ADI-58125 will be devised.
[0789] To establish safety margins, exposures anticipated for the proposed 1200 mg IM dose determined from the PopPK model described above were compared against exposures achieved in the
GLP rat toxicology study. These data suggest safety margins of ~ 3-fold and 51 -fold for AUC and Cmax, respectively, relative to the 1200 mg IM dose in humans, as shown in Table 20.
Table 20: Estimated Safety Margins for Proposed ADI-58125 Clinical Dose Based on Serum Exposures and Body Weight Extrapolation
Abbreviations: AUC=area under the serum concentration-time curve, AUCo 52weeks=AUC from time zero to 52 weeks post dose, AUC0-inf=AUC from time zero to infinity, Cmax=maximum serum concentration, IM=intramuscular; IV=intravenous, NOAEL=no observed adverse-effect level.* Doses evaluated in the FIH study. Note: Simulations presented in Table 20 are based on the PopPK model with a typical subject (weight = 80.9 kg). Safety margins were calculated by dividing the sex- combined rat mean Day 22 parameters at the NOAEL of 300 mg/kg (IV) (Cmax: 7890 μg/mL, AUCo inf: 1590000 hr*μg/mL) by the simulated human Cmax and AUCo 52 weeks. AUC-related safety margin calculations are based on AUC0-inf data.
[0790] Data generated to date across the clinical program also suggest that ADI-58125 had been well-tolerated across diverse study populations. No injection site reactions, hypersensitivity reactions, or other study-drug related AEs had been observed at any dose level in review of blinded safety data with a minimum of 9 months of follow-up. Similarly, ongoing internal review of blinded safety data from STAMP and EVADE has revealed no safety concerns. A single hypersensitivity event (urticaria/hives) has been reported, which was graded by the investigator as mild and was self-limited (did not require any treatment). The independent data monitoring committee (iDMC) has also reviewed unblinded data on three occasions, with a blinded safety update provided for EVADE, and recommended that the studies continue.
[0791] Based on enrollment to date, approximately 1460 participants had received ADI-58125 at dose levels ranging from 300 mg IM to 500 mg IV/600 mg IM across the clinical trials. Given the lack of identified safety signals to date, the adequate safety margins (based on AUC, Cmax, and body weight extrapolation), the lack of ADI-58125 -specific binding in human TCR studies, the lack of ADI-58125-specific findings in the rat GLP toxicology study, and the prospect for benefit in the setting of prevention in the face of the surging Omicron variant, the potential benefit of the 1200 mg IM dose outweighs the potential risk to participants and the proposed dose can be incorporated into the EVADE trial with safety oversight.
Treatment of SARS-CoV-2: ADI-58125 -TRMT-001 (STAMP)
[0792] The dose selection fof ADI-58125 for the treatment indication against the Omicron variant was evaluated and compared to prior variants, and it was concluded that both a higher dose and alternative administration route (intravenous) were needed to achieve desired exposures for treatment of disease resulting from this variant.
[0793] In order to identify the appropriate dosing regimen in the setting of treatment, the above mentioned PopPK model based on phase 1 data was used. Dose selection was designed to achieve a tissue-adjusted concentration exceeding the in vitro IC90 for the Omicron variant in 90% of simulated patients, evaluating ELF: serum ratios of 10% and 15%. A dose of 1200 mg administered IV is projected to achieve concentrations of ADI-58125 in serum associated with ELF concentrations at or above IC90 for Omicron as well as other SARS-CoV-2 VOI/VOCs through 28 days in 90% of simulated patients using an ELF: serum ratio of 10%. The IC90 value generated against authentic virus was used for dose selection to maintain consistency with prior dose selection efforts.
[0794] Exposures anticipated for the proposed 1200 mg IV dose level determined from the PopPK model and safety margins based on the GLP rat toxicology study are presented in Table 20. These data suggest safety margins of 2.3-fold and 26-fold for AUC and Cmax, respectively, relative to the 1200 mg IV dose in humans.
[0795] Based on enrollment to date, approximately 1460 participants had received ADG20 at dose levels ranging from 300 mg IM to 500 mg IV/600 mg IM across the clinical trials. Given the lack of identified safety signals to date, the adequate safety margins (based on AUC, Cmax, and body weight extrapolation), the lack of ADG20-specific binding in human TCR studies, the lack of ADG20- specific findings in the rat GLP toxicology study, and the prospect for benefit in the setting of treatment in the face of the surging Omicron variant, the potential benefit of the 1200 mg IV dose outweighs the potential risk to participants and that the proposed dose can be incorporated into the STAMP study with safety oversight.
Additional Omicron Neutralization Results
[0796] ADI-58125 activity against the Omicron variant were evaluated in multiple authentic and pseudotyped VLP neutralization assays across different laboratories (Table 21). As presented in Table 21, IC50 values against Omicron range from 0.129 to 1.703 μg/mL. For the purpose of dose selection, the IC50 and IC90 values were used to maintain consistency across prior dose selection efforts. ADI- 58125 neutralized authentic Omicron virus with an IC50 of 1.029 μg/mL and IC90 of 9.943 μg/mL, representing 257- and 663-fold reductions in activity relative to the Victoria reference strain, respectively. Furthermore, authorized mAbs were tested in parallel in the same assay. In addition to ADI-58125, only S309, AZD1061, and AZD8895, as well as the AZD7442 cocktail, achieved >50% neutralization within the antibody concentrations tested, and these mAbs displayed IC50 values
ranging from 0.319 to 2.03 mg/mL (Figure 48). However, AZD1061 and AZD8895 did not reach 90% neutralization, while S309 and AZD7442 exhibited IC90 values of 5.92 and 2.03 mg/mL, respectively. REGN10933, REGN10987, LY-C0VI6, and LY-CoV555 displayed complete or near-complete loss of activity. Across two pseudotyped lentiviral neutralization assays, ADI-58125 displayed 65- to 328- fold reduced potency against B.1.1.529/BA.1.
VSV=vesicular stomatitis virus a IC50 and IC90 values shown were separately calculated using raw percentage neutralization data from Dejnirattisai 2022. Neutralization curves were fitted using nonlinear regression with four- parameter variable slope to calculate the IC50. IC90 was determined by constraining parameter F (fraction of maximal response) to 90. All analyses were performed using GraphPad Prism (version 9.3.1).
[0797] Additional studies conducted using ADI-58125 and ADI-58124 were also summarized in Table 21. These included authentic and pseudotyped vesicular stomatitis virus (VSV) neutralization
assays that demonstrated 21- to 183-fold changes in IC50 compared to each study's respective reference strain. ADI-58125 and ADI-58124 activity had also been evaluated in pseudotyped viral- like particles (VLPs) expressing the full set of Omicron spike mutations plus an R346K mutation, which has been detected in a subset of circulating Omicron variants. Based on lentiviral and VSV pseudo virus neutralization data, ADI-58125 activity against Omicron+R346K appeared to be similar to its reported activity against Omicron (Table 21) (Liu 2021b). Detailed assay methodologies for each study are provided in Table 22.
References:
1. Abu-Raddad, L. J., H. Chemaitelly and A. A. Butt. (2021). Effectiveness of the BNT162b2 Covid-19 Vaccine against the B. 1.1. 7 and B. 1.351 Variants.
2. Andrews, N., J. Stowe, F. Kirsebom, et al. (2021). Effectiveness of COVID-19 vaccines against the Omicron (B. 1.1. 529) variant of concern.
3. AstraZeneca. (2021). Fact Sheet for Healthcare Providers: Emergency Use Authorization for EVUSHELD (tixagevimab co-packaged with cilgavimab).
4. Cameroni, E., C. Saliba, J. E. Bowen, et al. (2021). Broadly neutralizing antibodies overcome SARS-CoV-2 Omicron antigenic shift.
5. Clemens, S. A. C., P. M. Folegatti, K. R. Emary, et al. (2021). Efficacy of ChAdOxl nCoV-19 (AZD1222) vaccine against SARS-CoV-2 lineages circulating in Brazil.
6. Dejnirattisai, W., J. Huo, D. Zhou, et al. (2022). SARS-CoV-2 Omicron-B. EE 529 leads to widespread escape from neutralizing antibody responses.
7. Deng, R., A. P. Lee, M. Maia, et al. (2018). Pharmacokinetics of MHAA4549A, an anti-influenza A monoclonal antibody, in healthy subjects challenged with influenza A virus in a phase Ila randomized trial. Clinical pharmacokinetics 57(3): 367-377.
8. ECDC. (2021). Country Overview Report: Week 50, 2021.
9. EMA. (2021a). EMA issues advice on use of Lagevrio (molnupiravir) for the treatment of COVID-19.
10. EMA. (2021b). EMA issues advice on use of Paxlovid (PF-07321332 and ritonavir) for the treatment of COVID-19: rolling review starts in parallel.
11. Emary, K. R., T. Golubchik, P. K. Aley, et al. (2021). Efficacy of ChAdOxl nCoV-19 (AZD1222) vaccine against SARS-CoV-2 variant of concern 202012/01 (B. 1.1. 7): an exploratory analysis of a randomised controlled trial.
12. Falsey, A. R., M. E. Sobieszczyk, I. Hirsch, et al. (2021). Phase 3 safety and efficacy of AZD1222 (ChAdOxl nCoV-19) COVID-19 vaccine.
13. FDA. (2020). Emergency Use Authorization (EUA) for casirivimab and imdevimab. Center for Drug Evaluation and Research (CDER) Review. 14 December 2020.
14. Hitchings, M. D., O. T. Ranzani, M. Dorion, et al. (2021). Effectiveness of ChAdOxl vaccine in older adults during SARS-CoV-2 Gamma variant circulation in Sao Paulo.
15. Huang, Y., O. Borisov, J. J. Kee, et al. (2021). Calibration of two validated SARS-CoV-2 pseudo virus neutralization assays for COVID-19 vaccine evaluation. Scientific reports 11(1): 1- 13.
16. Liu, C., H. M. Ginn, W. Dejnirattisai, et al. (2021a, 2021/06/17/). Reduced neutralization of SARS-CoV-2 B.1.617 by vaccine and convalescent serum.
17. Liu, L., S. Iketani, Y. Guo, et al. (2021b). Striking Antibody Evasion Manifested by the Omicron Variant of SARS-CoV-2.
18. Loo, Y.-M., P. M. McTamney, R. H. Arends, et al. (2021). AZD7442 demonstrates prophylactic and therapeutic efficacy in non-human primates and extended half-life in humans. medRxiv.
19. Lopez Bernal, J., N. Andrews, C. Gower, et al. (2021). Effectiveness of Covid-19 vaccines against the B. 1.617. 2 (Delta) variant.
20. Madhi, S. A., V. Baillie, C. L. Cutland, et al. (2021, May 20). Efficacy of the ChAdOxl nCoV-19 Covid-19 Vaccine against the B.1.351 Variant. N Engl J Med 2021/03/17.
21. Nasreen, S., S. He, H. Chung, et al. (2021). Effectiveness of COVID-19 vaccines against variants of concern, Canada.
22. Neerukonda, S. N., R. Vassell, R. Herrup, et al. (2021). Establishment of a well-characterized SARS-CoV-2 lentiviral pseudovirus neutralization assay using 293T cells with stable expression of ACE2 and TMPRSS2. Plos one 16(3): e0248348.
23. O'Brien, M. P., E. F. Neto, B. J. Musser, et al. (2021). Subcutaneous REGEN-COV Antibody Combination for Covid-19 Prevention. medRxiv.
24. Planas, D., N. Saunders, P. Maes, et al. (2021). Considerable escape of SARS-CoV-2 variant Omicron to antibody neutralization.
25. Polack, F. P., S. J. Thomas, N. Kitchin, et al. (2020). Safety and Efficacy of the BNT162b2 mRNA Covid-19 Vaccine. N Engl J Med 383(27): 2603-2615.
26. Regeneron. (2021). New Phase 3 Analysis Show That a Single Dose of REGEN- COV®(Casirivimab and imdevimab) Provides Long-Term Protection Against COVID-19.
27. Tartof, S. Y., J. M. Slezak, H. Fischer, et al. (2021). Effectiveness of mRNA BNT162b2 COVID- 19 vaccine up to 6 months in a large integrated health system in the USA: a retrospective cohort study. The Lancet 398(10309): 1407-1416.
28. UIP. (2022). 12 cases of Omicron have been confirmed in Bulgaria after cellogenomic sequencing.
29. UKHSA. (2021). SARS-CoV-2 variants of concern and variants under investigation in England: Technical briefing 33.
30. UMOH. (2021). New cases of Omicron strain detected.
31. WHO. (2022). Ukraine: WHO Coronavirus Disease (COVID-19) Dashboard with Vaccination Data. Updated 04 January 2022. Retrieved 04 January 2022.
32. Wollacott, A. M., M. F. Boni, K. J. Szretter, et al. (2016). Safety and upper respiratory pharmacokinetics of the hemagglutinin stalk-binding antibody VIS410 support treatment and prophylaxis based on population modeling of seasonal influenza A outbreaks. EBioMedicine 5: 147-155.
Example 18. Preliminary Results from the Phase 2/3 Randomized, Double-Blind, Placebo- Controlled Trial to Evaluate the Efficacy and Safety of ADI-58125 in the Treatment of Ambulatory Participants with Mild or Moderate COVID-19 (STAMP)
[0798] The STAMP trial is evaluating the safety and efficacy of ADI-58125 as a potential treatment for mild or moderate COVID-19 in ambulatory patients with a high risk of disease progression based on age or comorbidities.
[0799] Patients were enrolled if they had a positive SARS-CoV-2 test and symptom duration of 5 days or less. Patients were randomized 1:1 to receive ADI-58125 or placebo administered by a single intramuscular (IM) injection and were followed through Day 29 for primary efficacy. The primary efficacy endpoint was COVID-19 related hospitalization or all-cause death through Day 29 in patients with disease due to confirmed or suspected SARS-CoV-2 variants other than Omicron.
[0800] In the primary efficacy analysis, which included 336 patients (n=169 ADI-58125, n=167 placebo), ADI-58125 was associated with a statistically significant reduction in the risk of COVID- 19-related hospitalization or all-cause death through Day 29 compared with placebo (8 [4.7%] vs. 23 [13.8%]), demonstrating a 66% relative risk reduction (RRR) in favor of ADI-58125. The standardized risk difference was -8.6% (95% Cl: -14.65, -2.57; p=0.0052). The favorable treatment effect for ADI-58125 was observed across key subgroups. Among patients treated within 3 days of symptom onset, ADI-58125 reduced the risk of COVID-19 hospitalization or all-cause death compared to placebo, with a standardized RRR of 75%. ADI-58125 provided a greater reduction in viral load from baseline to Day 5 compared with placebo as assessed by saliva samples, with an adjusted least-squares mean difference of -0.81 logio copies/mL (95% Cl: -1.3, -0.3; p=0.0020). The median follow-up for safety analyses was 99 days (n=192 ADI-58125, n=200 placebo). No study drug related SAEs, including deaths, and no hypersensitivity reactions were reported. The most frequently reported AEs were solicited injection site reactions, all of which were mild to moderate in severity and similar in incidence between arms.
[0801] A single dose of ADI-58125 300 mg IM provided a statistically significant and clinically meaningful reduction in the risk of COVID-19 related hospitalization and all-cause death compared to placebo in high-risk ambulatory patients with mild to moderate COVID-19. ADI-58125 was well tolerated with a safety profile similar to that of placebo.
Example 19. Clinical and Virologic Outcomes with Early ADI-58125 Monoclonal Antibody Therapy in Mild and Moderate COVID-19
[0802] Peak SARS-CoV-2 viral replication occurs in the upper respiratory tract in presymptomatic and early symptomatic phases. Administration of a monoclonal antibody may be most beneficial in the early time period immediately after symptom onset. Here the effect of early therapy on efficacy in patients receiving ADI-58125 was evaluated.
[0803] High risk patients with mild or moderate COVID-19 were enrolled in the ADI-58125 treatment study (STAMP), with primary endpoint of COVID-19 related hospitalization or all-cause death through Day 29 in patients with disease due to confirmed or suspected SARS-CoV-2 variants other than Omicron. Patients were randomized 1 : 1 to receive ADI or placebo administered by a single intramuscular (IM) injection. For this subgroup analysis, patients that had received therapy within 3 days of symptom onset were evaluated.
[0804] In the overall population, the study met the primary endpoint demonstrating 66% relative risk reduction of COVID-19 hospitalization or all cause death in 336 patients. Among 261 patients receiving therapy within 3 days of symptom onset (n=133 ADI-58125, n=128 placebo), ADI- 58125 was associated with a statistically significant reduction in the risk of COVID-19-related hospitalization or all-cause death through Day 29 compared with placebo (4 [3%] vs. 15 [11.7%], standardized risk difference -8%, 95% Cl: -14.11, -1.86, p=0.0106), demonstrating a 72% standardized relative risk reduction in favor of ADI-58125. When given as early therapy, ADI-58125 provided a greater reduction in viral load from baseline to Day 5 compared with placebo as assessed by saliva samples, with an adjusted least-squares mean difference of - 0.97 loglO copies/mL (95% Cl: -1.540, -0.391; p=0.0011). No study drug related SAEs, including deaths, and no hypersensitivity reactions were reported.
[0805] Early therapy with a single dose of ADI-58125 300 mg IM provided a 72% reduction in the risk of COVID-19 related hospitalization and all-cause death compared to placebo in high-risk ambulatory patients with mild to moderate COVID-19. Therapy within the first 3 days also led to a greater reduction in viral load compared to placebo and favorable outcomes in patients who are at high risk for progression of disease.
Example 20. Validation of a Predictive Model to Correlate Neutralization Titers and Efficacy for the Prevention of COVID-19
[0806] Most anti-viral vaccines, including COVID-19 vaccines, induce neutralizing antibodies as a correlate of protection (CoP). Here a novel model was decribed to predict the absolute serum neutralizing antibody titer required for prevention of symptomatic COVID-19, and its validation from Phase 2/3 clinical trial of ADI-58125 for prevention.
[0807] To determine neutralizing antibody (nAb) titers that were associated with protection against symptomatic COVID-19, a literature search was conducted to evaluate efficacy data from vaccines and monoclonal antibodies (mAh). Median authentic virus serum neutralizing titers induced by vaccination or passive mAB therapy were measured experimentally or projected based on mAh pharmacokinetic (PK) and neutralization IC50 data. From previous FIH study of ADI-58125, PK and neutralizing titers correlate in a predictable manner. Therefore, to validate the CoP, serum concentrations from population PK model based on Phase 2/3 data of ADI-58125 on days 55 and 76 (Omicron BA.1/BA1.1), and 90 (Delta) were used to calculate predictive nAb titers and corresponding efficacy for the Delta and Omicron variants and then compared to clinical efficacy results from the EVADE clinical trial in prevention of COVID-19.
[0808] A CoP was identified as an authentic virus serum neutralizing titer of 1 : 100 which would provide approximately 70% protection against development of symptomatic COVID-19. Population PK median serum concentrations of ADI-58125 at 55, 76, and 90 days were 29.8 mg/L, 26.7 mg/L, and 24.6 mg/L, respectively. The neutralizing titer against Delta was extrapolated to be 1:4100 at day 90, predicting ADI-58125 would have at least 70% protection. This was consistent with the observed clinical outcome from the EVADE trial where patients taking ADI-58125 experienced a 71% reduction in development of symptomatic COVID-19 through 3 months. Neutralizing titers against Omicron were extrapolated to be 1:29 and 1:26, at days 55 and 76, respectively. The model predicted an efficacy of approximately 50% and 46.6% for ADI-58125 against Omicron given a higher IC50. This was in range of the observed efficacy of 62.3% and 50.9% at a median follow-up of 55 and 76 days in EVADE (Figure 55).
[0809] In conclusion, an authentic virus serum neutralizing titer of 1 : 100 was established as a CoP for the prevention of symptomatic COVID-19 and validated using Phase 2/3 clinical data in a model that could be used to predict expected outcomes and redosing strategy based on IC50 of the circulating variant.
[0810] Having fully described and enabled the invention, the invention is further described by the claims that follow. In general, in the following claims, the terms used should not be construed to limit the disclosure to the specific embodiments disclosed in the specification and the claims. Accordingly,
the invention is not limited by the disclosure, but instead the scope of the invention is to be determined entirely by the following claims.
[0811] The disclosure may be practiced in ways other than those particularly described in the foregoing description and examples. Numerous modifications and variations of the disclosure are possible in light of the above teachings and, therefore, are within the scope of the appended claims.
Claims
1. A method of inducing an immune response against a coronavirus (CoV) in a subject in need thereof, the method comprising administering to the subject an isolated antibody, or antigen-binding fragment thereof, which binds to the spike protein of a coronavirus (“CoV-S”), wherein said antibody, or antigen-binding fragment thereof, comprises a heavy chain variable region (VH) comprising a VH CDR1 comprising SEQ ID NO:52, a VH CDR2 comprising SEQ ID NO:54, and a VH CDR3 comprising SEQ ID NO:56, and a light chain variable region (VL) comprising a VL CDR1 comprising SEQ ID NO:252, a VL CDR2 comprising SEQ ID NO:254, and a VL CDR3 comprising SEQ ID NO:256;
(a) wherein the antibody, or antigen-binding fragment thereof, reaches a maximum concentration (Cmax) of about 10 μg/mL to about 1000 μg/mL, about 20 μg/mL to about 500 μg/mL, about 30 μg/mL to about 400 μg/mL, about 40 μg/mL to about 300 μg/mL, about 50 μg/mL to about 200 μg/mL, about 50 μg/mL to about 100 μg/mL, or about 30 μg/mL to about 70 μg/mL, about 100 μg/mL to about 400 μg/mL, about 150 μg/mL to about 350 μg/mL in serum of the subject;
(b) wherein the median time (Tmax) for the antibody, or antigen-binding fragment thereof, to reach a maximum concentration in serum of the subject is about 0.01-30 days, about 3-20 days, about 6-20 days, about 7-18 days, about 8-15 or about 13-15 days;
(c) wherein the antibody, or antigen-binding fragment thereof, reaches a maximum concentration of about 30 μg/mL to about 200 μg/mL, about 100 μg/mL to about 200 μg/mL, about 30 μg/mL to about 100 μg/mL, about 40 μg/mL to about 80 μg/mL, about 50 μg/mL to about 70 μg/mL, or about 30 μg/mL to about 65 μg/mL in serum of the subject in about 0.01-20 days, about 3-20 days, about 6-20 days, about 7-18 days, about 8-15 or about 13-15 days after administration;
(d) wherein the area under the serum concentration-time curve from day 0 to day 21 (AUC0 -21 d) is about 100-2000 day* μg/mL, about 1000-2000 day* μg/mL, about 1400-2000 day* μg/mL, about 200-1500 day* μg/mL, about 400-1400 day* μg/mL, about 500-1300 day* μg/mL, about 600- 1000 day* μg/mL, or about 800-900 day* μg/mL;
(e) wherein the area under the serum concentration-time curve from day 0 to day 90 (AUC0 -90 d) is about 1000-10000 day* μg/mL, about 2000-8000 day* μg/mL, about 2000-5000 day* μg/mL, about 3000-4000 day* μg/mL, about 5000-10000 day* μg/mL, about 2000-4000 day* μg/mL, about 5000-8000 day* μg/mL, or about 6000-8000 day* μg/mL;
(f) wherein the area under the serum concentration-time curve from day 0 to day 180 (AUC0 -180d) is about 1000-10000 day* μg/mL, about 2000-5000 day* μg/mL, about 3000-6000 day* μg/mL, about 5000-10000 day* μg/mL, about 5000-8000 day* μg/mL, or about 4000-6000 day* μg/mL;
(g) wherein the area under the serum concentration-time curve from day 0 to day 365 (AUC0-365d) is about 1000-30000 day* mg/mL, about 5000-30000 day* mg/mL, about 10000-30000 day* mg/mL, about 10000-15000 day* mg/mL, about 20000-30000 day* mg/mL, or about 25000-30000 day* mg/mL; and/or
(h) wherein the antibody, or antigen-binding fragment thereof, has a serum half-life of about 50-200 days, about 50-160 days, about 50-140 days, about 40-130 days, about 50-100 days, about 60-100 days, about 60-90 days, about 70-90 days, about 90-100 days, about 70-80 days, about 80-130 days, about 100-150 days, about 110-140 days, or about 120-140 days.
2. A method of preventing a coronavirus infection in a subject in need thereof, the method comprising administering to the subject an isolated antibody, or antigen-binding fragment thereof, which binds to the spike protein of a coronavirus (“CoV-S”), wherein said antibody, or antigen-binding fragment thereof, comprises a heavy chain variable region (VH) comprising a VH CDR1 comprising SEQ ID NO:52, a VH CDR2 comprising SEQ ID NO:54, and a VH CDR3 comprising SEQ ID NO:56, and a light chain variable region (VL) comprising a VL CDR1 comprising SEQ ID NO:252, a VL CDR2 comprising SEQ ID NO:254, and a VL CDR3 comprising SEQ ID NO:256;
(a) wherein the antibody, or antigen-binding fragment thereof, reaches a maximum concentration (Cmax) of about 10 μg/mL to about 1000 μg/mL, about 20 μg/mL to about 500 μg/mL, about 30 μg/mL to about 400 μg/mL, about 40 μg/mL to about 300 μg/mL, about 50 μg/mL to about 200 μg/mL, about 50 μg/mL to about 100 μg/mL, or about 30 μg/mL to about 70 μg/mL, about 100 μg/mL to about 400 μg/mL, about 150 μg/mL to about 350 μg/mL in serum of the subject;
(b) wherein the median time (Tmax) for the antibody, or antigen-binding fragment thereof, to reach a maximum concentration in serum of the subject is about 0.01-30 days, about 3-20 days, about 6-20 days, about 7-18 days, about 8-15 or about 13-15 days;
(c) wherein the antibody, or antigen-binding fragment thereof, reaches a maximum concentration of about 30 μg/mL to about 200 μg/mL, about 100 μg/mL to about 200 μg/mL, about 30 μg/mL to about 100 μg/mL, about 40 μg/mL to about 80 μg/mL, about 50 μg/mL to about 70 μg/mL, or about 30 μg/mL to about 65 μg/mL in serum of the subject in about 0.01-20 days, about 3-20 days, about 6-20 days, about 7-18 days, about 8-15 or about 13-15 days after administration;
(d) wherein the area under the serum concentration-time curve from day 0 to day 21 (AUC0 -21 d) is about 100-2000 day* μg/mL, about 1000-2000 day* μg/mL, about 1400-2000 day* μg/mL, about 200-1500 day* μg/mL, about 400-1400 day* μg/mL, about 500-1300 day* μg/mL, about 600- 1000 day* μg/mL, or about 800-900 day* μg/mL;
(e) wherein the area under the serum concentration-time curve from day 0 to day 90 (AUC0 -90 d) is about 1000-10000 day* μg/mL, about 2000-8000 day* μg/mL, about 2000-5000 day* μg/mL,
about 3000-4000 day* mg/mL, about 5000-10000 day* mg/mL, about 2000-4000 day* mg/mL, about 5000-8000 day* mg/mL, or about 6000-8000 day* mg/mL;
(f) wherein the area under the serum concentration-time curve from day 0 to day 180 (§UC0 -180d) is about 1000-10000 day* mg/mL, about 2000-5000 day* mg/mL, about 3000-6000 day* mg/mL, about 5000-10000 day* mg/mL, about 5000-8000 day* mg/mL, or about 4000-6000 day* mg/mL;
(g) wherein the area under the serum concentration-time curve from day 0 to day 365 (AUC0-365d) is about 1000-30000 day* mg/mL, about 5000-30000 day* mg/mL, about 10000-30000 day* mg/mL, about 10000-15000 day* mg/mL, about 20000-30000 day* mg/mL, or about 25000-30000 day* mg/mL; and/or
(h) wherein the antibody, or antigen-binding fragment thereof, has a serum half-life of about 50-200 days, about 50-160 days, about 50-140 days, about 40-130 days, about 50-100 days, about 60-100 days, about 60-90 days, about 70-90 days, about 90-100 days, about 70-80 days, about 80-130 days, about 100-150 days, about 110-140 days, or about 120-140 days.
3. A method of treating a coronavirus infection in a subject in need thereof, the method comprising administering to the subject an isolated antibody, or antigen-binding fragment thereof, which binds to the spike protein of a coronavirus (“CoV-S”), wherein said antibody, or antigen-binding fragment thereof, comprises a heavy chain variable region (VH) comprising a VH CDR1 comprising SEQ ID NO:52, a VH CDR2 comprising SEQ ID NO:54, and a VH CDR3 comprising SEQ ID NO:56, and a light chain variable region (VL) comprising a VL CDR1 comprising SEQ ID NO:252, a VL CDR2 comprising SEQ ID NO:254, and a VL CDR3 comprising SEQ ID NO:256;
(a) wherein the antibody, or antigen-binding fragment thereof, reaches a maximum concentration (Cmax) of about 10 μg/mL to about 1000 μg/mL, about 20 μg/mL to about 500 μg/mL, about 30 μg/mL to about 400 μg/mL, about 40 μg/mL to about 300 μg/mL, about 50 μg/mL to about 200 μg/mL, about 50 μg/mL to about 100 μg/mL, or about 30 μg/mL to about 70 μg/mL, about 100 μg/mL to about 400 μg/mL, about 150 μg/mL to about 350 μg/mL in serum of the subject;
(b) wherein the median time (Tmax) for the antibody, or antigen-binding fragment thereof, to reach a maximum concentration in serum of the subject is about 0.01-30 days, about 3-20 days, about 6-20 days, about 7-18 days, about 8-15 or about 13-15 days;
(c) wherein the antibody, or antigen-binding fragment thereof, reaches a maximum concentration of about 30 μg/mL to about 200 μg/mL, about 100 μg/mL to about 200 μg/mL, about 30 μg/mL to about 100 μg/mL, about 40 μg/mL to about 80 μg/mL, about 50 μg/mL to about 70 μg/mL, or about 30 μg/mL to about 65 μg/mL in serum of the subject in about 0.01-20 days, about 3-20 days, about 6-20 days, about 7-18 days, about 8-15 or about 13-15 days after administration;
(d) wherein the area under the serum concentration-time curve from day 0 to day 21 (AUC0 -21 d) is about 100-2000 day* mg/mL, about 1000-2000 day* mg/mL, about 1400-2000 day* mg/mL, about 200-1500 day* mg/mL, about 400-1400 day* mg/mL, about 500-1300 day* mg/mL, about 600- 1000 day* mg/mL, or about 800-900 day* mg/mL;
(e) wherein the area under the serum concentration-time curve from day 0 to day 90 (AUC0 -90 d) is about 1000-10000 day* mg/mL, about 2000-8000 day* mg/mL, about 2000-5000 day* mg/mL, about 3000-4000 day* mg/mL, about 5000-10000 day* mg/mL, about 2000-4000 day* mg/mL, about 5000-8000 day* mg/mL, or about 6000-8000 day* mg/mL;
(f) wherein the area under the serum concentration-time curve from day 0 to day 180 (AUC0 -180d) is about 1000-10000 day* mg/mL, about 2000-5000 day* mg/mL, about 3000-6000 day* mg/mL, about 5000-10000 day* mg/mL, about 5000-8000 day* mg/mL, or about 4000-6000 day* mg/mL;
(g) wherein the area under the serum concentration-time curve from day 0 to day 365 (AUC0-365d) is about 1000-30000 day* mg/mL, about 5000-30000 day* mg/mL, about 10000-30000 day* mg/mL, about 10000-15000 day* mg/mL, about 20000-30000 day* mg/mL, or about 25000-30000 day* mg/mL; and/or
(h) wherein the antibody, or antigen-binding fragment thereof, has a serum half-life of about 50-200 days, about 50-160 days, about 50-140 days, about 40-130 days, about 50-100 days, about 60-100 days, about 60-90 days, about 70-90 days, about 90-100 days, about 70-80 days, about 80-130 days, about 100-150 days, about 110-140 days, or about 120-140 days.
4. A method of treating a symptom of a coronavirus infection in a subject in need thereof, the method comprising administering to the subject an isolated antibody, or antigen-binding fragment thereof, which binds to the spike protein of a coronavirus (“CoV-S”), wherein said antibody, or antigen-binding fragment thereof, comprises a heavy chain variable region (VH) comprising a VH CDR1 comprising SEQ ID NO:52, a VH CDR2 comprising SEQ ID NO:54, and a VH CDR3 comprising SEQ ID NO:56, and a light chain variable region (VL) comprising a VL CDR1 comprising SEQ ID NO:252, a VL CDR2 comprising SEQ ID NO:254, and a VL CDR3 comprising SEQ ID NO:256;
(a) wherein the antibody, or antigen-binding fragment thereof, reaches a maximum concentration (Cmax) of about 10 μg/mL to about 1000 μg/mL, about 20 μg/mL to about 500 μg/mL, about 30 μg/mL to about 400 μg/mL, about 40 μg/mL to about 300 μg/mL, about 50 μg/mL to about 200 μg/mL, about 50 μg/mL to about 100 μg/mL, or about 30 μg/mL to about 70 μg/mL, about 100 μg/mL to about 400 μg/mL, about 150 μg/mL to about 350 μg/mL in serum of the subject;
(b) wherein the median time (Tmax) for the antibody, or antigen-binding fragment thereof, to reach a maximum concentration in serum of the subject is about 0.01-30 days, about 3-20 days, about 6-20 days, about 7-18 days, about 8-15 or about 13-15 days;
(c) wherein the antibody, or antigen-binding fragment thereof, reaches a maximum concentration of about 30 mg/mL to about 200 mg/mL, about 100 mg/mL to about 200 mg/mL, about 30 mg/mL to about 100 mg/mL, about 40 mg/mL to about 80 mg/mL, about 50 mg/mL to about 70 mg/mL, or about 30 mg/mL to about 65 mg/mL in serum of the subject in about 0.01-20 days, about 3-20 days, about 6-20 days, about 7-18 days, about 8-15 or about 13-15 days after administration;
(d) wherein the area under the serum concentration-time curve from day 0 to day 21 (AUC0 -21 d) is about 100-2000 day* mg/mL, about 1000-2000 day* mg/mL, about 1400-2000 day* mg/mL, about 200-1500 day* mg/mL, about 400-1400 day* mg/mL, about 500-1300 day* mg/mL, about 600- 1000 day* mg/mL, or about 800-900 day* mg/mL;
(e) wherein the area under the serum concentration-time curve from day 0 to day 90 (AUC0 -90 d) is about 1000-10000 day* mg/mL, about 2000-8000 day* mg/mL, about 2000-5000 day* mg/mL, about 3000-4000 day* mg/mL, about 5000-10000 day* mg/mL, about 2000-4000 day* mg/mL, about 5000-8000 day* mg/mL, or about 6000-8000 day* mg/mL;
(f) wherein the area under the serum concentration-time curve from day 0 to day 180 (AUC0 -180d) is about 1000-10000 day* mg/mL, about 2000-5000 day* mg/mL, about 3000-6000 day* mg/mL, about 5000-10000 day* mg/mL, about 5000-8000 day* mg/mL, or about 4000-6000 day* mg/mL;
(g) wherein the area under the serum concentration-time curve from day 0 to day 365 (AUC0-365d) is about 1000-30000 day* mg/mL, about 5000-30000 day* mg/mL, about 10000-30000 day* mg/mL, about 10000-15000 day* mg/mL, about 20000-30000 day* mg/mL, or about 25000-30000 day* mg/mL; and/or
(h) wherein the antibody, or antigen-binding fragment thereof, has a serum half-life of about 50-200 days, about 50-160 days, about 50-140 days, about 40-130 days, about 50-100 days, about 60-100 days, about 60-90 days, about 70-90 days, about 90-100 days, about 70-80 days, about 80-130 days, about 100-150 days, about 110-140 days, or about 120-140 days.
5. The method of claim 4, wherein the symptom comprises at least one of bronchitis, pneumonia, respiratory failure, acute respiratory failure, organ failure, multi-organ system failure, pediatric inflammatory multisystem syndrome, acute respiratory distress syndrome, blood clot, a cardiac condition, myocardial injury, myocarditis, heart failure, cardiac arrest, acute myocardial infarction, dysrhythmia, venous thromboembolism, post-intensive care syndrome, shock, anaphylactic shock, cytokine release syndrome, septic shock, disseminated intravascular coagulation, ischemic stroke, intracerebral hemorrhage, microangiopathic thrombosis, psychosis, seizure, nonconvulsive status epilepticus, traumatic brain injury, stroke, anoxic brain injury, encephalitis, posterior reversible leukoencephalopathy, necrotizing encephalopathy, post-infectious encephalitis, autoimmune mediated encephalitis, acute disseminated encephalomyelitis, acute kidney injury, acute liver injury, pancreatic injury, immune thrombocytopenia, subacute thyroiditis, a gastrointestinal complication, aspergillosis,
increased susceptibility to infection with another virus or bacteria, and/or a pregnancy-related complication.
6. A method of decreasing the risk of mortality, hospitalization, mechanical ventilation, or a combination thereof, in a subject infected by a coronavirus, the method comprising administering to the subject an isolated antibody, or antigen-binding fragment thereof, which binds to the spike protein of a coronavirus (“CoV-S”), wherein said antibody, or antigen-binding fragment thereof, comprises a heavy chain variable region (VH) comprising a VH CDR1 comprising SEQ ID NO:52, a VH CDR2 comprising SEQ ID NO:54, and a VH CDR3 comprising SEQ ID NO:56, and a light chain variable region (VL) comprising a VL CDR1 comprising SEQ ID NO:252, a VL CDR2 comprising SEQ ID NO:254, and a VL CDR3 comprising SEQ ID NO:256;
(a) wherein the antibody, or antigen-binding fragment thereof, reaches a maximum concentration (Cmax) of about 10 μg/mL to about 1000 μg/mL, about 20 μg/mL to about 500 μg/mL, about 30 μg/mL to about 400 μg/mL, about 40 μg/mL to about 300 μg/mL, about 50 μg/mL to about 200 μg/mL, about 50 μg/mL to about 100 μg/mL, or about 30 μg/mL to about 70 μg/mL, about 100 μg/mL to about 400 μg/mL, about 150 μg/mL to about 350 μg/mL in serum of the subject;
(b) wherein the median time (Tmax) for the antibody, or antigen-binding fragment thereof, to reach a maximum concentration in serum of the subject is about 0.01-30 days, about 3-20 days, about 6-20 days, about 7-18 days, about 8-15 or about 13-15 days;
(c) wherein the antibody, or antigen-binding fragment thereof, reaches a maximum concentration of about 30 μg/mL to about 200 μg/mL, about 100 μg/mL to about 200 μg/mL, about 30 μg/mL to about 100 μg/mL, about 40 μg/mL to about 80 μg/mL, about 50 μg/mL to about 70 μg/mL, or about 30 μg/mL to about 65 μg/mL in serum of the subject in about 0.01-20 days, about 3-20 days, about 6-20 days, about 7-18 days, about 8-15 or about 13-15 days after administration;
(d) wherein the area under the serum concentration-time curve from day 0 to day 21 (AUC0 -21 d) is about 100-2000 day* μg/mL, about 1000-2000 day* μg/mL, about 1400-2000 day* μg/mL, about 200-1500 day* μg/mL, about 400-1400 day* μg/mL, about 500-1300 day* μg/mL, about 600- 1000 day* μg/mL, or about 800-900 day* μg/mL;
(e) wherein the area under the serum concentration-time curve from day 0 to day 90 (AUC0 -90 d) is about 1000-10000 day* μg/mL, about 2000-8000 day* μg/mL, about 2000-5000 day* μg/mL, about 3000-4000 day* μg/mL, about 5000-10000 day* μg/mL, about 2000-4000 day* μg/mL, about 5000-8000 day* μg/mL, or about 6000-8000 day* μg/mL;
(f) wherein the area under the serum concentration-time curve from day 0 to day 180 (AUC0 -180d) is about 1000-10000 day* μg/mL, about 2000-5000 day* μg/mL, about 3000-6000 day* μg/mL, about 5000-10000 day* μg/mL, about 5000-8000 day* μg/mL, or about 4000-6000 day* μg/mL;
(g) wherein the area under the serum concentration-time curve from day 0 to day 365 (AUCo 365d) is about 1000-30000 day* mg/mL, about 5000-30000 day* mg/mL, about 10000-30000 day* mg/mL, about 10000-15000 day* mg/mL, about 20000-30000 day* mg/mL, or about 25000-30000 day* mg/mL; and/or
(h) wherein the antibody, or antigen-binding fragment thereof, has a serum half-life of about 50-200 days, about 50-160 days, about 50-140 days, about 40-130 days, about 50-100 days, about 60-100 days, about 60-90 days, about 70-90 days, about 90-100 days, about 70-80 days, about 80-130 days, about 100-150 days, about 110-140 days, or about 120-140 days.
7. The method of any one of claims 1-6, wherein the CoV-S is the spike protein of SARS-CoV (“SARS-CoV-S”) or the spike protein of SARS-CoV-2 (“SARS-CoV-2-S”).
8. The method of any one of claims 1-7, wherein the antibody, or antigen-binding fragment thereof, cross-reacts with SARS-CoV-S and SARS-CoV -2-S.
9. The method of any one of claims 1-8, wherein SARS-CoV-S comprises an amino acid sequence of SEQ ID NO: 401, and wherein SARS-CoV-2-S comprises an amino acid sequence of SEQ ID NO: 403.
10. The method of any one of claims 1-9, wherein the SARS-CoV -2-S is a B.1.1.7 variant, a B. 1.351 variant, a B.1.1.28 variant, a B. 1.429 variant, a P.l variant, a B.1.617 variant, a B.1.617.2 variant, a C.37 variant, a 1.621 variant, a AY.l variant, a 1.623 variant, a C.36 variant, a A.27 variant, a AV.l variant, a B.1.1.482 variant, a B.1.1.523 variant, a B.1.427 variant, a AY.4 variant, a AY.ll variant, a D614G variant of SEQ ID NO: 403, a B.1.1.529/BA.1 variant, a BA.1.1 variant, or a BA.2 variant.
11. A method for inducing an immune response against a coronavirus Omicron variant in a subject in need thereof, for treating a coronavirus infection caused by an Omicron variant in a subject in need thereof, for preventing a coronavirus infection caused by an Omicron variant in a subject in need thereof, for treating a symptom of an infection of a subject by an Omicron variant in a subject in need thereof, and/or for decreasing the risk of mortality, hospitalization, mechanical ventilation, or a combination thereof, in a subject infected by an Omicron variant, the method comprising administering to the subject an isolated antibody, or antigen-binding fragment thereof, wherein said antibody, or antigen-binding fragment thereof, comprises a heavy chain variable region (VH) comprising a VH CDR1 comprising SEQ ID NO:52, a VH CDR2 comprising SEQ ID NO:54, and a VH CDR3 comprising SEQ ID NO:56, and a light chain variable region (VL)
comprising a VL CDR1 comprising SEQ ID NO:252, a VL CDR2 comprising SEQ ID NO:254, and a VL CDR3 comprising SEQ ID NO:256.
12. The method of claim 11, wherein the Omicron variant comprises a B.1.1.529/BA.1 variant, a BA.1.1 variant, or a BA.2 variant.
13. The method of any one of claims 1-12, wherein the VH comprises a sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to SEQ ID NO:58 and the VL comprises a sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to SEQ ID NO:258.
14. The method of any one of claims 1-13, wherein the VH comprises or consists of SEQ ID NO:58 and the VL comprises or consists of SEQ ID NO:258.
15. The method of any one of claims 1-14, wherein the subject is a human subject.
16. The method of any one of claims 1-15, wherein the subject is an adult, an adolescent, or a child.
17. The method of any one of claims 1-16, wherein the subject has at least one risk factor which renders them more prone to a poor clinical outcome.
18. The method of claim 17, wherein the at least one risk factor is selected from the group consisting of: an old age selected from the group consisting of over 55, over 60 or over 65 years old; diabetes, a chronic respiratory condition, obesity, hypertension, a cardiac or cardiovascular condition, a chronic inflammatory or autoimmune condition, and an immune compromised status.
19. The method of any one of claims 1-18, wherein the subject is immunocompromised.
20. The method of any one of claims 1-19, wherein the subject is at a high risk of disease progression.
21. The method of claim 20, wherein the subject is (a) age 50 years or above with no comorbid conditions or additional risk factors for progression of COVID-19; (b) between about 18 to about 50 years old and with one or more preexisting medical conditions selected from the group consisting of obesity, diabetes, chronic kidney disease, chronic lung disease, cardiac disease, sickle cell disease or thalassemia, solid organ or blood stem cell transplant recipient, other immunodeficiency due to
underlying illness or immunosuppressant medication, Down Syndrome, stroke or cerebrovascular disease, substance use disorder, and pregnancy; or (c) between about 12 to about 17 years old and with one or more preexisting medical conditions selected from the group consisting of obesity, diabetes, chronic kidney disease, sickle cell disease or thalassemia, congenital or acquired heart disease, neurodevelopmental disorder, a medically-related technological dependence, asthma or chronic respiratory disease, solid organ or blood stem cell transplant recipient, other immunodeficiency due to underlying illness or immunosuppressant medication, Down Syndrome, stroke or cerebrovascular disease, substance use disorder, and pregnancy.
22. The method of claim 20 or 21, wherein the subject is age 50 or above with no comorbid conditions or additional risk factors for progression of COVID-19.
23. The method of claim 20 or 21 , wherein the subject has hypertension with at least one medication prescribed or recommended.
24. The method of claim 20 or 21, wherein the subject has moderate to severe asthma requiring daily therapy.
25. The method of any one of claims 1-24, wherein the antibody, or the antigen-binding fragment thereof, is administered intramuscularly or intravenously.
26. The method of any one of claims 1-25, wherein the antibody, or the antigen-binding fragment thereof, is administered at a dose of about 100 mg to about 5000 mg, about 300 mg to about 4500 mg, about 500 mg to about 4500 mg, about 1200 mg to about 4500 mg, about 100 mg to about 2000 mg, about 200 mg to about 1500 mg, about 300 mg to about 600 mg, about 500 mg to about 1200 mg, about 300 mg to about 1200 mg; or at a dose of about 4500 mg, a dose of about 1200 mg, a dose of about 600 mg, or a dose of about 500 mg; or as two doses of 600 mg each on the same day, or as two doses of 300 mg each on the same day.
27. The method of any one of claims 1-26, wherein the antibody, or antigen-binding fragment thereof, is administered once, or is administered weekly.
28. The method of any one of claims 1-27, wherein the method further comprises obtaining an epithelial lining fluid (ELF) sample from the subject.
29. The method of any one of claims 1-28, wherein the ELF sample comprises an ELF sample from an upper airway, an lower airway, and/or an alveolar tissue.
30. The method of any one of claims 1-29, wherein the antibody, or antigen-binding fragment thereof, reaches a concentration of about 1 μg/mL to about 100 μg/mL, about 1 μg/mL to about 80 μg/mL, about 80 μg/mL to about 100 μg/mL, about 50 μg/mL to about 100 μg/mL, about 1 μg/mL to about 50 μg/mL, about 2 μg/mL to about 25 μg/mL, or about 2 μg/mL to about 10 μg/mL, in the ELF sample of the subject.
31. The method of any one of claims 1-30, wherein the antibody, or antigen-binding fragment thereof, has a receptor occupancy of at least 50%, 60%, 70%, 80%, or 90% in the ELF sample.
32. The method of any one of claims 1-31, wherein the antibody, or antigen-binding fragment thereof, has a receptor occupancy of at least 50%, 60%, 70%, 80%, or 90% in the ELF sample for about 28 days after administration.
33. The method of any one of claims 1-32, wherein the antibody, or antigen-binding fragment thereof, has a virus neutralizing titer of about 100-2000, about 200-1500, about 300-1500, or about 500- 1500 in serum of the subject about 6 months after administration.
34. The method of any one of claims 1-33, wherein a 80% virus neutralization titer (MN80) of the antibody, or antigen-binding fragment thereof, at about days 0-14, about days 7-21, about 3 months, or about 6 months after administration, is about 10-6000, about 50-600, about 500-1500, about 1000-2500, about 100-2500, about 500-2000, about 500-1500, about 400-1200, about 200-1500, about 300-1000, about 400-800, about 400-1000, or about 500-600 in the subject.
35. The method of claim 34, wherein the 80% virus neutralization titer (MN80) of the antibody, or antigen-binding fragment thereof, at about day 7, day 14, or day 21 after administration, is about 300- 2000, about 400-600, about 600-1500, about 1100-1700, about 500-1700, about 500-1500, about 400- 1200, about 400-800, about 400-1000, or about 500-600 in the serum sample of the subject.
36. The method of claim 34, wherein the 80% virus neutralization titer (MN80) of the antibody, or antigen-binding fragment thereof, at about 3 months after administration, is about 200-1000, about 200- 800, about 200-500, about 400-900, or about 400-600 in the serum sample of the subject.
37. The method of claim 34, wherein the 80% virus neutralization titer (MN80) of the antibody, or antigen-binding fragment thereof, at about 6 months after administration, is about 10-500, about 300- 500, or about 50-200 in the serum sample of the subject.
38. The method of any one of claims 1-37, wherein a 50% virus neutralization titer (MN50) of the antibody, or antigen-binding fragment thereof, at about days 0-14, about days 7-21, about 3 months, about 6 months, or about 12 months after administration, is about 100-6000, about 300-1500, about 1700-3800, about 3800-5200, about 300-5500, about 1200-4500, about 1300-4300, about 1200-4000, about 100-2500, about 500-2500, about 800-2000, about 1000-1800, about 800-1300, about 900-1100, or about 1300-1500 in the serum sample of the subject.
39. The method of claim 38, wherein the 50% virus neutralization titer (MN50) of the antibody, or antigen-binding fragment thereof, at about day 7, day 14 or day 21 after administration, is about 1000- 4500, about 1200-4500, about 1300-4300, about 1200-3900, about 1500-4000, about 1800-3800, about 3800-4500, about 1000-1800 or about 1200-1500 in the serum sample of the subject.
40. The method of claim 38, wherein the 50% virus neutralization titer (MN50) of the antibody, or antigen-binding fragment thereof, at about 3 months after administration, is about 800-1300, or about 900-1100 in the serum sample of the subject.
41. The method of claim 38, wherein the 50% virus neutralization titer (MN50) of the antibody, or antigen-binding fragment thereof, at about 6 months after administration, is about 200-500, or about 300-600 in the serum sample of the subject.
42. The method of claim 38, wherein the 50% virus neutralization titer (MN50) of the antibody, or antigen-binding fragment thereof, at about 12 months after administration, is about 150-500, or about 200-400 in the serum sample of the subject.
43. The method of any one of claims 33-42, wherein the virus neutralization titer is determined using a plaque reduction neutralization test (PRNT).
44. The method of any one of claims 1-43, wherein the serum concentration of the antibody, or antigen-binding fragment thereof, is about 1-300 mg/L, about 1-250 mg/L, about 1-200 mg/L, about 1- 100 mg/L, about 100-250 mg/L, about 150-200 mg/L, about 120-170 mg/L, about 1-90 mg/L, about 1- 80 mg/L, about 1-70 mg/L, about 1-60 mg/L, about 1-50 mg/L, about 1-40 mg/L, about 1-30 mg/L,
about 1-20 mg/L, about 1-10 mg/L, about 2-9 mg/L, about 3-8 mg/L, about 4-6 mg/L, about 5-80 mg/L, about 10-70 mg/L, or about 10-60 mg/L in the subject about 3 months after administration.
45. The method of any one of claims 1-44, wherein the serum concentration of the antibody, or antigen-binding fragment thereof, is about 1-100 mg/L, about 1-90 mg/L, about 1-80 mg/L, about 1-70 mg/L, about 1-60 mg/L, about 1-50 mg/L, about 1-40 mg/L, about 1-30 mg/L, about 1-20 mg/L, about 1-10 mg/L, about 2-9 mg/L, about 3-8 mg/L, about 4-6 mg/L, about 5-80 mg/L, about 10-70 mg/L, about 10-60 mg/L, about 5-25 mg/L, about 10-30 mg/L, about 15-25 mg/L, about 20-100 mg/L, about 30-100 mg/L, about 50-80 mg/L, or about 60-70 mg/L in the subject about 6 months after administration.
46. The method of any one of claims 1-45, wherein the serum concentration of the antibody, or antigen-binding fragment thereof, is about 0.1-30 mg/L, about 1-30 mg/L, about 1-20 mg/L, about 1- 10 mg/L, about 10-20 mg/L, about 0.1-3 mg/L, about 0.5-8 mg/L, about 0.5-10 mg/L, about 2-9 mg/L, about 5-15 mg/L, about 3-8 mg/L, or about 4-6 mg/L in the subject about 12 months after administration.
47. The method of any one of claims 1-46, wherein the clearance rate for the antibody, or antigen binding fragment thereof, is about 0.1-10 mL/d, about 0.1-5.0 mL/d, about 0.1-3.0 mL/d, about 0.5-2.5 mL/d, or about 1.0-2.0 mL/d.
48. The method of any one of claims 1-47, wherein the steady state volume of distribution for the antibody, or antigen-binding fragment thereof, is about 1-10 L, about 2-8 L, about 4-9 L, about 4-8 L, or about 5-7L.
49. The method of any one of claims 1-48, wherein the antibody, or antigen-binding fragment thereof, is administered in combination with at least one antibody selected from the group consisting of ADI-58120, ADI-58121, ADI-58122, ADI-58123, ADI-58124, ADI-58125, ADI-58126, ADI-58127, ADI-58128, ADI-58129, ADI-58130 and ADI-58131.
50. The method of any one of claims 1-49, wherein the antibody, or antigen-binding fragment thereof, is administered in combination with a vaccine.
51. The method of claim 50, wherein the antibody, or antigen-binding fragment thereof, is administered (a) concurrently with a COVID-19 vaccine; (b) after administration of a COVID-19 vaccine, or (c) prior to administration of a COVID-19 vaccine.
52. The method of any one of claims 1-51, wherein administration of the antibody, or antigen binding fragment thereof, reduces pulmonary inflammation in the subject.
53. The method of any one of claims 1-52, wherein administration of the antibody, or antigen binding fragment thereof, reduces the risk of COVID-19 hospitalization or death of the subject.
54. The method of any one of claims 1-53, wherein administration of the antibody, or antigen binding fragment thereof, reduces viral load in the subject.
55. The method of any one of claims 1-54, wherein the antibody, or antigen-binding fragment thereof, is administered to the subject within 5, 4, 3, 2 or 1 days of symptom onset.
Applications Claiming Priority (40)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US202163182033P | 2021-04-30 | 2021-04-30 | |
US63/182,033 | 2021-04-30 | ||
US202163191410P | 2021-05-21 | 2021-05-21 | |
US63/191,410 | 2021-05-21 | ||
US202163197717P | 2021-06-07 | 2021-06-07 | |
US63/197,717 | 2021-06-07 | ||
US202163211334P | 2021-06-16 | 2021-06-16 | |
US63/211,334 | 2021-06-16 | ||
US202163233443P | 2021-08-16 | 2021-08-16 | |
US63/233,443 | 2021-08-16 | ||
US202163243259P | 2021-09-13 | 2021-09-13 | |
US63/243,259 | 2021-09-13 | ||
US202163245509P | 2021-09-17 | 2021-09-17 | |
US63/245,509 | 2021-09-17 | ||
US202163249158P | 2021-09-28 | 2021-09-28 | |
US63/249,158 | 2021-09-28 | ||
US202163256753P | 2021-10-18 | 2021-10-18 | |
US63/256,753 | 2021-10-18 | ||
US202163279202P | 2021-11-15 | 2021-11-15 | |
US63/279,202 | 2021-11-15 | ||
US202163281914P | 2021-11-22 | 2021-11-22 | |
US63/281,914 | 2021-11-22 | ||
US202163283433P | 2021-11-27 | 2021-11-27 | |
US63/283,433 | 2021-11-27 | ||
US202163292781P | 2021-12-22 | 2021-12-22 | |
US63/292,781 | 2021-12-22 | ||
US202263298832P | 2022-01-12 | 2022-01-12 | |
US63/298,832 | 2022-01-12 | ||
US202263302714P | 2022-01-25 | 2022-01-25 | |
US63/302,714 | 2022-01-25 | ||
US202263316067P | 2022-03-03 | 2022-03-03 | |
US63/316,067 | 2022-03-03 | ||
US202263324569P | 2022-03-28 | 2022-03-28 | |
US63/324,569 | 2022-03-28 | ||
US202263324995P | 2022-03-29 | 2022-03-29 | |
US63/324,995 | 2022-03-29 | ||
US202263327436P | 2022-04-05 | 2022-04-05 | |
US63/327,436 | 2022-04-05 | ||
US202263332312P | 2022-04-19 | 2022-04-19 | |
US63/332,312 | 2022-04-19 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2022232557A1 true WO2022232557A1 (en) | 2022-11-03 |
Family
ID=83848858
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2022/027004 WO2022232557A1 (en) | 2021-04-30 | 2022-04-29 | Compounds specific to coronavirus s protein and uses thereof |
Country Status (2)
Country | Link |
---|---|
TW (1) | TW202309075A (en) |
WO (1) | WO2022232557A1 (en) |
Citations (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2007044695A2 (en) * | 2005-10-07 | 2007-04-19 | Dana-Farber Cancer Institute | ANTIBODIES AGAINST SARS-CoV AND METHODS OF USE THEREOF |
US20090311265A1 (en) * | 2006-09-07 | 2009-12-17 | Van Den Brink Edward Norbert | Human binding molecules capable or neutralizing influenze virus h5n1 and uses thereof |
US20130302318A1 (en) * | 2010-09-27 | 2013-11-14 | Lisa Rojkjaer | Anti-CD38 Antibody and Lenalidomide or Bortezomib for the Treatment of Multipe Myeloma and NHL |
US20130330335A1 (en) * | 2010-03-23 | 2013-12-12 | Iogenetics, Llc | Bioinformatic processes for determination of peptide binding |
-
2022
- 2022-04-29 TW TW111116513A patent/TW202309075A/en unknown
- 2022-04-29 WO PCT/US2022/027004 patent/WO2022232557A1/en active Application Filing
Patent Citations (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2007044695A2 (en) * | 2005-10-07 | 2007-04-19 | Dana-Farber Cancer Institute | ANTIBODIES AGAINST SARS-CoV AND METHODS OF USE THEREOF |
US20090311265A1 (en) * | 2006-09-07 | 2009-12-17 | Van Den Brink Edward Norbert | Human binding molecules capable or neutralizing influenze virus h5n1 and uses thereof |
US20130330335A1 (en) * | 2010-03-23 | 2013-12-12 | Iogenetics, Llc | Bioinformatic processes for determination of peptide binding |
US20130302318A1 (en) * | 2010-09-27 | 2013-11-14 | Lisa Rojkjaer | Anti-CD38 Antibody and Lenalidomide or Bortezomib for the Treatment of Multipe Myeloma and NHL |
Also Published As
Publication number | Publication date |
---|---|
TW202309075A (en) | 2023-03-01 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
US11220536B1 (en) | Compounds specific to coronavirus S protein and uses thereof | |
AU2019370339B2 (en) | Bispecific antibody and use thereof | |
AU2020286284B2 (en) | Novel anti-CD39 antibodies | |
WO2022170126A2 (en) | Compounds specific to coronavirus s protein and uses thereof | |
AU2014233478A1 (en) | Anti-CD25 antibodies and their uses | |
TWI778985B (en) | Anti-chikv antibodies and uses thereof | |
JP2023080074A (en) | Anti-trem-1 antibodies and uses thereof | |
TR201810773T4 (en) | Oncostatin (osm) antigen binding proteins. | |
JP2018519832A (en) | Multispecific binding protein | |
US20220010003A1 (en) | Anti-periostin antibodies and uses thereof | |
JP2019511532A (en) | Methods of treating inflammatory diseases | |
US11359007B2 (en) | Anti-SARS-CoV-2 neutralizing antibodies | |
WO2022232557A1 (en) | Compounds specific to coronavirus s protein and uses thereof | |
WO2024050356A2 (en) | Sars-cov2 antibodies and uses thereof | |
CA3120063A1 (en) | Flt3 agonist antibodies and uses thereof | |
WO2023193017A2 (en) | Sars-cov2 antibodies and uses thereof | |
WO2023059900A2 (en) | Pharmaceutical formulations comprising anti-coronavirus s protein antibodies and uses thereof | |
KR20230058057A (en) | antigen binding protein | |
WO2022160013A1 (en) | Sars-cov-2 binding proteins | |
CA3102329A1 (en) | Novel anti-cd39 antibodies |
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: 22796838 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: 22796838 Country of ref document: EP Kind code of ref document: A1 |