WO2024245527A1 - Treatment of pneumonia and ards with inhibitors of c5a and il-6 activity - Google Patents
Treatment of pneumonia and ards with inhibitors of c5a and il-6 activity Download PDFInfo
- Publication number
- WO2024245527A1 WO2024245527A1 PCT/EP2023/064290 EP2023064290W WO2024245527A1 WO 2024245527 A1 WO2024245527 A1 WO 2024245527A1 EP 2023064290 W EP2023064290 W EP 2023064290W WO 2024245527 A1 WO2024245527 A1 WO 2024245527A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- activity
- inhibitor
- seq
- sequence according
- tocilizumab
- Prior art date
Links
- 230000000694 effects Effects 0.000 title claims abstract description 322
- 239000003112 inhibitor Substances 0.000 title claims abstract description 298
- 238000011282 treatment Methods 0.000 title claims abstract description 145
- 206010035664 Pneumonia Diseases 0.000 title claims abstract description 33
- 102100031506 Complement C5 Human genes 0.000 claims abstract description 242
- 102000004889 Interleukin-6 Human genes 0.000 claims abstract description 166
- 108090001005 Interleukin-6 Proteins 0.000 claims abstract description 166
- 208000015181 infectious disease Diseases 0.000 claims abstract description 47
- 230000002458 infectious effect Effects 0.000 claims abstract description 45
- 206010001052 Acute respiratory distress syndrome Diseases 0.000 claims abstract description 36
- 229960003989 tocilizumab Drugs 0.000 claims description 137
- 229940062471 vilobelimab Drugs 0.000 claims description 115
- 230000027455 binding Effects 0.000 claims description 94
- 230000006698 induction Effects 0.000 claims description 52
- 239000000427 antigen Substances 0.000 claims description 49
- 108091007433 antigens Proteins 0.000 claims description 49
- 102000036639 antigens Human genes 0.000 claims description 49
- 239000012634 fragment Substances 0.000 claims description 49
- 108090000623 proteins and genes Proteins 0.000 claims description 37
- 235000018102 proteins Nutrition 0.000 claims description 36
- 102000004169 proteins and genes Human genes 0.000 claims description 36
- 208000025721 COVID-19 Diseases 0.000 claims description 35
- 235000001014 amino acid Nutrition 0.000 claims description 34
- 102000010781 Interleukin-6 Receptors Human genes 0.000 claims description 32
- 108010038501 Interleukin-6 Receptors Proteins 0.000 claims description 32
- 150000001413 amino acids Chemical class 0.000 claims description 32
- 238000012423 maintenance Methods 0.000 claims description 31
- 239000003446 ligand Substances 0.000 claims description 30
- 108010059426 Anaphylatoxin C5a Receptor Proteins 0.000 claims description 26
- 102000005590 Anaphylatoxin C5a Receptor Human genes 0.000 claims description 26
- 102100035360 Cerebellar degeneration-related antigen 1 Human genes 0.000 claims description 26
- 125000003275 alpha amino acid group Chemical group 0.000 claims description 21
- 101100112922 Candida albicans CDR3 gene Proteins 0.000 claims description 20
- 101000737793 Homo sapiens Cerebellar degeneration-related antigen 1 Proteins 0.000 claims description 19
- 102100035361 Cerebellar degeneration-related protein 2 Human genes 0.000 claims description 18
- 101000737796 Homo sapiens Cerebellar degeneration-related protein 2 Proteins 0.000 claims description 18
- 208000013616 Respiratory Distress Syndrome Diseases 0.000 claims description 14
- 201000000028 adult respiratory distress syndrome Diseases 0.000 claims description 14
- 206010035737 Pneumonia viral Diseases 0.000 claims description 8
- 208000009421 viral pneumonia Diseases 0.000 claims description 8
- FWMNVWWHGCHHJJ-SKKKGAJSSA-N 4-amino-1-[(2r)-6-amino-2-[[(2r)-2-[[(2r)-2-[[(2r)-2-amino-3-phenylpropanoyl]amino]-3-phenylpropanoyl]amino]-4-methylpentanoyl]amino]hexanoyl]piperidine-4-carboxylic acid Chemical compound C([C@H](C(=O)N[C@H](CC(C)C)C(=O)N[C@H](CCCCN)C(=O)N1CCC(N)(CC1)C(O)=O)NC(=O)[C@H](N)CC=1C=CC=CC=1)C1=CC=CC=C1 FWMNVWWHGCHHJJ-SKKKGAJSSA-N 0.000 claims description 7
- 150000003384 small molecules Chemical class 0.000 claims description 7
- 230000003612 virological effect Effects 0.000 claims description 7
- 230000001419 dependent effect Effects 0.000 claims description 6
- 108010054477 Immunoglobulin Fab Fragments Proteins 0.000 claims description 5
- 102000001706 Immunoglobulin Fab Fragments Human genes 0.000 claims description 5
- 230000002401 inhibitory effect Effects 0.000 claims description 5
- 230000009870 specific binding Effects 0.000 claims description 5
- 108010003723 Single-Domain Antibodies Proteins 0.000 claims description 4
- 229950002507 elsilimomab Drugs 0.000 claims description 4
- 229940121578 levilimab Drugs 0.000 claims description 4
- 229950010006 olokizumab Drugs 0.000 claims description 4
- 229950006348 sarilumab Drugs 0.000 claims description 4
- 229960003323 siltuximab Drugs 0.000 claims description 4
- 229950006094 sirukumab Drugs 0.000 claims description 4
- 201000001178 Bacterial Pneumonia Diseases 0.000 claims description 3
- 108700022150 Designed Ankyrin Repeat Proteins Proteins 0.000 claims description 3
- 238000012867 alanine scanning Methods 0.000 claims description 3
- 230000001580 bacterial effect Effects 0.000 claims description 3
- 230000000903 blocking effect Effects 0.000 claims description 3
- 229940126681 complement 5a receptor antagonist Drugs 0.000 claims description 3
- 229950007269 vobarilizumab Drugs 0.000 claims description 3
- 230000002538 fungal effect Effects 0.000 claims description 2
- 230000004071 biological effect Effects 0.000 claims 1
- 229940100601 interleukin-6 Drugs 0.000 description 136
- 241000282414 Homo sapiens Species 0.000 description 51
- 238000004458 analytical method Methods 0.000 description 46
- 239000000902 placebo Substances 0.000 description 29
- 229940068196 placebo Drugs 0.000 description 29
- 238000000034 method Methods 0.000 description 24
- 210000004027 cell Anatomy 0.000 description 22
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 22
- 239000003814 drug Substances 0.000 description 22
- 229940126602 investigational medicinal product Drugs 0.000 description 21
- -1 IL-ip Proteins 0.000 description 18
- 108060003951 Immunoglobulin Proteins 0.000 description 18
- 102000018358 immunoglobulin Human genes 0.000 description 18
- 108090000765 processed proteins & peptides Proteins 0.000 description 18
- 230000004083 survival effect Effects 0.000 description 18
- 108010047041 Complementarity Determining Regions Proteins 0.000 description 17
- 150000001875 compounds Chemical class 0.000 description 17
- 229940079593 drug Drugs 0.000 description 17
- 230000003993 interaction Effects 0.000 description 16
- 102000005962 receptors Human genes 0.000 description 14
- 108020003175 receptors Proteins 0.000 description 14
- 239000000243 solution Substances 0.000 description 14
- 201000010099 disease Diseases 0.000 description 13
- 238000001802 infusion Methods 0.000 description 13
- 210000000440 neutrophil Anatomy 0.000 description 12
- 229910052760 oxygen Inorganic materials 0.000 description 12
- 241001465754 Metazoa Species 0.000 description 11
- 229910002092 carbon dioxide Inorganic materials 0.000 description 11
- 125000001072 heteroaryl group Chemical group 0.000 description 11
- 238000007619 statistical method Methods 0.000 description 11
- 210000004072 lung Anatomy 0.000 description 10
- 238000002483 medication Methods 0.000 description 10
- 102000004196 processed proteins & peptides Human genes 0.000 description 10
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 9
- 125000003118 aryl group Chemical group 0.000 description 9
- 208000035475 disorder Diseases 0.000 description 9
- 230000009467 reduction Effects 0.000 description 9
- 230000002829 reductive effect Effects 0.000 description 9
- 241000894007 species Species 0.000 description 9
- 229910052717 sulfur Inorganic materials 0.000 description 9
- 238000010494 dissociation reaction Methods 0.000 description 8
- 230000005593 dissociations Effects 0.000 description 8
- 125000005842 heteroatom Chemical group 0.000 description 8
- 239000000203 mixture Substances 0.000 description 8
- 238000012360 testing method Methods 0.000 description 8
- 208000001528 Coronaviridae Infections Diseases 0.000 description 7
- 125000000217 alkyl group Chemical group 0.000 description 7
- 238000011161 development Methods 0.000 description 7
- 230000018109 developmental process Effects 0.000 description 7
- 229910052736 halogen Inorganic materials 0.000 description 7
- 150000002367 halogens Chemical group 0.000 description 7
- 229910052739 hydrogen Inorganic materials 0.000 description 7
- 239000001257 hydrogen Substances 0.000 description 7
- 125000004435 hydrogen atom Chemical group [H]* 0.000 description 7
- 239000012528 membrane Substances 0.000 description 7
- 229910052757 nitrogen Inorganic materials 0.000 description 7
- 238000012216 screening Methods 0.000 description 7
- 238000013517 stratification Methods 0.000 description 7
- 238000006467 substitution reaction Methods 0.000 description 7
- 125000004209 (C1-C8) alkyl group Chemical group 0.000 description 6
- 125000006648 (C1-C8) haloalkyl group Chemical group 0.000 description 6
- 102100032996 C5a anaphylatoxin chemotactic receptor 2 Human genes 0.000 description 6
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 6
- 101000868001 Homo sapiens C5a anaphylatoxin chemotactic receptor 2 Proteins 0.000 description 6
- 206010061218 Inflammation Diseases 0.000 description 6
- 230000004913 activation Effects 0.000 description 6
- 125000000539 amino acid group Chemical group 0.000 description 6
- 230000034994 death Effects 0.000 description 6
- 125000004404 heteroalkyl group Chemical group 0.000 description 6
- 210000004408 hybridoma Anatomy 0.000 description 6
- 230000004054 inflammatory process Effects 0.000 description 6
- 125000004433 nitrogen atom Chemical group N* 0.000 description 6
- 229920001184 polypeptide Polymers 0.000 description 6
- 238000002560 therapeutic procedure Methods 0.000 description 6
- 230000009261 transgenic effect Effects 0.000 description 6
- 125000005913 (C3-C6) cycloalkyl group Chemical group 0.000 description 5
- 241001678559 COVID-19 virus Species 0.000 description 5
- 101100012887 Drosophila melanogaster btl gene Proteins 0.000 description 5
- 238000002965 ELISA Methods 0.000 description 5
- 208000004852 Lung Injury Diseases 0.000 description 5
- 229910003827 NRaRb Inorganic materials 0.000 description 5
- 201000003176 Severe Acute Respiratory Syndrome Diseases 0.000 description 5
- 206010069363 Traumatic lung injury Diseases 0.000 description 5
- 238000006243 chemical reaction Methods 0.000 description 5
- 230000006378 damage Effects 0.000 description 5
- 125000000592 heterocycloalkyl group Chemical group 0.000 description 5
- 230000006872 improvement Effects 0.000 description 5
- 239000011780 sodium chloride Substances 0.000 description 5
- 208000024891 symptom Diseases 0.000 description 5
- CURLTUGMZLYLDI-UHFFFAOYSA-N Carbon dioxide Chemical compound O=C=O CURLTUGMZLYLDI-UHFFFAOYSA-N 0.000 description 4
- 102000004127 Cytokines Human genes 0.000 description 4
- 108090000695 Cytokines Proteins 0.000 description 4
- 206010014522 Embolism venous Diseases 0.000 description 4
- 208000025370 Middle East respiratory syndrome Diseases 0.000 description 4
- 241000577979 Peromyscus spicilegus Species 0.000 description 4
- 208000004756 Respiratory Insufficiency Diseases 0.000 description 4
- 241000700605 Viruses Species 0.000 description 4
- 230000009471 action Effects 0.000 description 4
- 239000005557 antagonist Substances 0.000 description 4
- QVGXLLKOCUKJST-UHFFFAOYSA-N atomic oxygen Chemical compound [O] QVGXLLKOCUKJST-UHFFFAOYSA-N 0.000 description 4
- 210000003719 b-lymphocyte Anatomy 0.000 description 4
- 230000008901 benefit Effects 0.000 description 4
- 230000015572 biosynthetic process Effects 0.000 description 4
- 238000011284 combination treatment Methods 0.000 description 4
- 230000024203 complement activation Effects 0.000 description 4
- 239000002552 dosage form Substances 0.000 description 4
- 238000013504 emergency use authorization Methods 0.000 description 4
- 230000006870 function Effects 0.000 description 4
- 230000036541 health Effects 0.000 description 4
- 238000002347 injection Methods 0.000 description 4
- 239000007924 injection Substances 0.000 description 4
- 231100000515 lung injury Toxicity 0.000 description 4
- 238000004519 manufacturing process Methods 0.000 description 4
- 230000007246 mechanism Effects 0.000 description 4
- 239000001301 oxygen Substances 0.000 description 4
- 230000037361 pathway Effects 0.000 description 4
- 238000011321 prophylaxis Methods 0.000 description 4
- 201000004193 respiratory failure Diseases 0.000 description 4
- 206010039073 rheumatoid arthritis Diseases 0.000 description 4
- 125000001424 substituent group Chemical group 0.000 description 4
- 229940124597 therapeutic agent Drugs 0.000 description 4
- 208000004043 venous thromboembolism Diseases 0.000 description 4
- 241000283707 Capra Species 0.000 description 3
- 102000004190 Enzymes Human genes 0.000 description 3
- 108090000790 Enzymes Proteins 0.000 description 3
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 3
- 241000233866 Fungi Species 0.000 description 3
- DHMQDGOQFOQNFH-UHFFFAOYSA-N Glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 description 3
- 101001076408 Homo sapiens Interleukin-6 Proteins 0.000 description 3
- 241000124008 Mammalia Species 0.000 description 3
- 241000699666 Mus <mouse, genus> Species 0.000 description 3
- 241000315672 SARS coronavirus Species 0.000 description 3
- 206010040047 Sepsis Diseases 0.000 description 3
- ZMANZCXQSJIPKH-UHFFFAOYSA-N Triethylamine Chemical compound CCN(CC)CC ZMANZCXQSJIPKH-UHFFFAOYSA-N 0.000 description 3
- 125000001931 aliphatic group Chemical group 0.000 description 3
- 125000003282 alkyl amino group Chemical group 0.000 description 3
- 125000004429 atom Chemical group 0.000 description 3
- 230000008827 biological function Effects 0.000 description 3
- 210000004369 blood Anatomy 0.000 description 3
- 239000008280 blood Substances 0.000 description 3
- 239000002975 chemoattractant Substances 0.000 description 3
- 210000004978 chinese hamster ovary cell Anatomy 0.000 description 3
- 229940124301 concurrent medication Drugs 0.000 description 3
- 125000004122 cyclic group Chemical group 0.000 description 3
- 125000004663 dialkyl amino group Chemical group 0.000 description 3
- 238000005516 engineering process Methods 0.000 description 3
- 230000007717 exclusion Effects 0.000 description 3
- 102000052611 human IL6 Human genes 0.000 description 3
- 210000000987 immune system Anatomy 0.000 description 3
- 230000001965 increasing effect Effects 0.000 description 3
- 210000000265 leukocyte Anatomy 0.000 description 3
- 230000000670 limiting effect Effects 0.000 description 3
- 125000005647 linker group Chemical group 0.000 description 3
- 210000004185 liver Anatomy 0.000 description 3
- 238000007477 logistic regression Methods 0.000 description 3
- 125000002496 methyl group Chemical group [H]C([H])([H])* 0.000 description 3
- 210000000056 organ Anatomy 0.000 description 3
- 238000006213 oxygenation reaction Methods 0.000 description 3
- 230000003285 pharmacodynamic effect Effects 0.000 description 3
- 238000002360 preparation method Methods 0.000 description 3
- 125000002924 primary amino group Chemical group [H]N([H])* 0.000 description 3
- 238000012959 renal replacement therapy Methods 0.000 description 3
- 150000003839 salts Chemical group 0.000 description 3
- 208000026425 severe pneumonia Diseases 0.000 description 3
- 230000011664 signaling Effects 0.000 description 3
- 238000002198 surface plasmon resonance spectroscopy Methods 0.000 description 3
- 210000001519 tissue Anatomy 0.000 description 3
- 238000002627 tracheal intubation Methods 0.000 description 3
- 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
- VBICKXHEKHSIBG-UHFFFAOYSA-N 1-monostearoylglycerol Chemical compound CCCCCCCCCCCCCCCCCC(=O)OCC(O)CO VBICKXHEKHSIBG-UHFFFAOYSA-N 0.000 description 2
- 241000894006 Bacteria Species 0.000 description 2
- 241000283690 Bos taurus Species 0.000 description 2
- 241000282836 Camelus dromedarius Species 0.000 description 2
- 241000711573 Coronaviridae Species 0.000 description 2
- 108020004414 DNA Proteins 0.000 description 2
- 101100012878 Drosophila melanogaster htl gene Proteins 0.000 description 2
- 108060006698 EGF receptor Proteins 0.000 description 2
- 241000196324 Embryophyta Species 0.000 description 2
- 241000283074 Equus asinus Species 0.000 description 2
- 241000283073 Equus caballus Species 0.000 description 2
- LYCAIKOWRPUZTN-UHFFFAOYSA-N Ethylene glycol Chemical compound OCCO LYCAIKOWRPUZTN-UHFFFAOYSA-N 0.000 description 2
- 241000282326 Felis catus Species 0.000 description 2
- 241000287828 Gallus gallus Species 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
- 108090000288 Glycoproteins Proteins 0.000 description 2
- 102000003886 Glycoproteins Human genes 0.000 description 2
- 108010001336 Horseradish Peroxidase Proteins 0.000 description 2
- 241001502974 Human gammaherpesvirus 8 Species 0.000 description 2
- 241000725303 Human immunodeficiency virus Species 0.000 description 2
- 108010021625 Immunoglobulin Fragments Proteins 0.000 description 2
- 102000008394 Immunoglobulin Fragments Human genes 0.000 description 2
- 108700005091 Immunoglobulin Genes Proteins 0.000 description 2
- 102000006496 Immunoglobulin Heavy Chains Human genes 0.000 description 2
- 108010019476 Immunoglobulin Heavy Chains Proteins 0.000 description 2
- 108010067060 Immunoglobulin Variable Region Proteins 0.000 description 2
- 102000017727 Immunoglobulin Variable Region Human genes 0.000 description 2
- 102000009571 Macrophage Inflammatory Proteins Human genes 0.000 description 2
- 108010009474 Macrophage Inflammatory Proteins Proteins 0.000 description 2
- 241000127282 Middle East respiratory syndrome-related coronavirus Species 0.000 description 2
- 241001529936 Murinae Species 0.000 description 2
- 108091028043 Nucleic acid sequence Proteins 0.000 description 2
- 241000283973 Oryctolagus cuniculus Species 0.000 description 2
- 241001494479 Pecora Species 0.000 description 2
- 241000009328 Perro Species 0.000 description 2
- 206010037660 Pyrexia Diseases 0.000 description 2
- 208000037847 SARS-CoV-2-infection Diseases 0.000 description 2
- 241000282898 Sus scrofa Species 0.000 description 2
- 210000001744 T-lymphocyte Anatomy 0.000 description 2
- 108060008682 Tumor Necrosis Factor Proteins 0.000 description 2
- 102100040247 Tumor necrosis factor Human genes 0.000 description 2
- 208000027418 Wounds and injury Diseases 0.000 description 2
- 230000002159 abnormal effect Effects 0.000 description 2
- 229940119059 actemra Drugs 0.000 description 2
- 230000003213 activating effect Effects 0.000 description 2
- 230000002411 adverse Effects 0.000 description 2
- 238000010171 animal model Methods 0.000 description 2
- 230000000890 antigenic effect Effects 0.000 description 2
- 238000003556 assay Methods 0.000 description 2
- GNNALEGJVYVIIH-UHFFFAOYSA-N benzene-1,2-diamine;hydrochloride Chemical compound Cl.NC1=CC=CC=C1N GNNALEGJVYVIIH-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
- 102000023732 binding proteins Human genes 0.000 description 2
- 108091008324 binding proteins Proteins 0.000 description 2
- 238000012575 bio-layer interferometry Methods 0.000 description 2
- 238000004364 calculation method Methods 0.000 description 2
- 239000001569 carbon dioxide Substances 0.000 description 2
- 125000003178 carboxy group Chemical group [H]OC(*)=O 0.000 description 2
- 239000000969 carrier Substances 0.000 description 2
- 230000001413 cellular effect Effects 0.000 description 2
- 230000008859 change Effects 0.000 description 2
- 239000003795 chemical substances by application Substances 0.000 description 2
- 230000003399 chemotactic effect Effects 0.000 description 2
- 229950001565 clazakizumab Drugs 0.000 description 2
- 230000000295 complement effect Effects 0.000 description 2
- 230000004154 complement system Effects 0.000 description 2
- 238000005094 computer simulation Methods 0.000 description 2
- 239000012141 concentrate Substances 0.000 description 2
- DDRJAANPRJIHGJ-UHFFFAOYSA-N creatinine Chemical compound CN1CC(=O)NC1=N DDRJAANPRJIHGJ-UHFFFAOYSA-N 0.000 description 2
- 238000013461 design Methods 0.000 description 2
- 239000003937 drug carrier Substances 0.000 description 2
- 238000001378 electrochemiluminescence detection Methods 0.000 description 2
- 239000012530 fluid Substances 0.000 description 2
- 238000009472 formulation Methods 0.000 description 2
- 108020001507 fusion proteins Proteins 0.000 description 2
- 102000037865 fusion proteins Human genes 0.000 description 2
- 210000004602 germ cell Anatomy 0.000 description 2
- 239000011521 glass Substances 0.000 description 2
- 230000011132 hemopoiesis Effects 0.000 description 2
- 230000002209 hydrophobic effect Effects 0.000 description 2
- 208000032641 idiopathic multicentric Castleman disease Diseases 0.000 description 2
- 210000002865 immune cell Anatomy 0.000 description 2
- 230000001900 immune effect Effects 0.000 description 2
- 230000028993 immune response Effects 0.000 description 2
- 229940072221 immunoglobulins Drugs 0.000 description 2
- 230000001939 inductive effect Effects 0.000 description 2
- 208000027866 inflammatory disease Diseases 0.000 description 2
- 230000005764 inhibitory process Effects 0.000 description 2
- 208000014674 injury Diseases 0.000 description 2
- 238000001990 intravenous administration Methods 0.000 description 2
- 238000000111 isothermal titration calorimetry Methods 0.000 description 2
- 108010045069 keyhole-limpet hemocyanin Proteins 0.000 description 2
- 239000007788 liquid Substances 0.000 description 2
- 239000008297 liquid dosage form Substances 0.000 description 2
- 231100000516 lung damage Toxicity 0.000 description 2
- 239000008176 lyophilized powder Substances 0.000 description 2
- 229920002521 macromolecule Polymers 0.000 description 2
- 210000002540 macrophage Anatomy 0.000 description 2
- 238000005259 measurement Methods 0.000 description 2
- 238000005399 mechanical ventilation Methods 0.000 description 2
- 230000001404 mediated effect Effects 0.000 description 2
- 244000005700 microbiome Species 0.000 description 2
- 210000001616 monocyte Anatomy 0.000 description 2
- 210000000066 myeloid cell Anatomy 0.000 description 2
- 230000007935 neutral effect Effects 0.000 description 2
- 229940021182 non-steroidal anti-inflammatory drug Drugs 0.000 description 2
- 238000002823 phage display Methods 0.000 description 2
- 239000000546 pharmaceutical excipient Substances 0.000 description 2
- 239000000244 polyoxyethylene sorbitan monooleate Substances 0.000 description 2
- 235000010482 polyoxyethylene sorbitan monooleate Nutrition 0.000 description 2
- 229920000053 polysorbate 80 Polymers 0.000 description 2
- 229940068968 polysorbate 80 Drugs 0.000 description 2
- 238000009597 pregnancy test Methods 0.000 description 2
- 230000001737 promoting effect Effects 0.000 description 2
- 238000003127 radioimmunoassay Methods 0.000 description 2
- 230000001105 regulatory effect Effects 0.000 description 2
- 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 2
- 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 2
- 238000011160 research Methods 0.000 description 2
- 238000012552 review Methods 0.000 description 2
- 239000001488 sodium phosphate Substances 0.000 description 2
- 229910000162 sodium phosphate Inorganic materials 0.000 description 2
- 239000000126 substance Substances 0.000 description 2
- 230000002195 synergetic effect Effects 0.000 description 2
- 230000000451 tissue damage Effects 0.000 description 2
- RYFMWSXOAZQYPI-UHFFFAOYSA-K trisodium phosphate Chemical compound [Na+].[Na+].[Na+].[O-]P([O-])([O-])=O RYFMWSXOAZQYPI-UHFFFAOYSA-K 0.000 description 2
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 2
- FPGZSNZMIGAHES-UCGGBYDDSA-N (2R,3S)-1-(2-fluoro-6-methylbenzoyl)-2-[4-(2-hydroxy-2-methylpropoxy)phenyl]-N-[4-methyl-3-(trifluoromethyl)phenyl]-3,4-dihydro-2H-quinoline-3-carboxamide Chemical compound CC1=C(C=C(NC(=O)[C@H]2CC3=C(C=CC=C3)N([C@H]2C2=CC=C(OCC(C)(C)O)C=C2)C(=O)C2=C(C)C=CC=C2F)C=C1)C(F)(F)F FPGZSNZMIGAHES-UCGGBYDDSA-N 0.000 description 1
- BPWYUCDDEBSUBZ-SLZJPVGVSA-N (2R,3S,5R)-2-[4-[(1-deuteriocyclopentyl)amino]phenyl]-1-(2,6-difluorobenzoyl)-N-[4-methyl-3-(trifluoromethyl)phenyl]-5-(trifluoromethyl)piperidine-3-carboxamide Chemical compound C1(CCCC1)([2H])NC1=CC=C(C=C1)[C@@H]1N(C[C@@H](C[C@@H]1C(=O)NC1=CC(=C(C=C1)C)C(F)(F)F)C(F)(F)F)C(C1=C(C=CC=C1F)F)=O BPWYUCDDEBSUBZ-SLZJPVGVSA-N 0.000 description 1
- 125000000229 (C1-C4)alkoxy group Chemical group 0.000 description 1
- 125000006552 (C3-C8) cycloalkyl group Chemical group 0.000 description 1
- OPRQHQNKHNUWJI-UHFFFAOYSA-N 1-(2,6-difluorobenzoyl)-5-(trifluoromethyl)piperidine-3-carboxamide Chemical compound FC1=C(C(=O)N2CC(CC(C2)C(F)(F)F)C(=O)N)C(=CC=C1)F OPRQHQNKHNUWJI-UHFFFAOYSA-N 0.000 description 1
- NMICZKAJOJLIRB-UHFFFAOYSA-N 1-(trifluoromethyl)piperidine-3-carboxamide Chemical compound NC(=O)C1CCCN(C(F)(F)F)C1 NMICZKAJOJLIRB-UHFFFAOYSA-N 0.000 description 1
- NCYCYZXNIZJOKI-IOUUIBBYSA-N 11-cis-retinal Chemical compound O=C/C=C(\C)/C=C\C=C(/C)\C=C\C1=C(C)CCCC1(C)C NCYCYZXNIZJOKI-IOUUIBBYSA-N 0.000 description 1
- MIJDSYMOBYNHOT-UHFFFAOYSA-N 2-(ethylamino)ethanol Chemical compound CCNCCO MIJDSYMOBYNHOT-UHFFFAOYSA-N 0.000 description 1
- 108010093667 ALX-0061 Proteins 0.000 description 1
- 206010069754 Acquired gene mutation Diseases 0.000 description 1
- 208000009304 Acute Kidney Injury Diseases 0.000 description 1
- 206010048998 Acute phase reaction Diseases 0.000 description 1
- 230000007730 Akt signaling Effects 0.000 description 1
- 108010088751 Albumins Proteins 0.000 description 1
- 102000009027 Albumins Human genes 0.000 description 1
- GUBGYTABKSRVRQ-XLOQQCSPSA-N Alpha-Lactose Chemical compound O[C@@H]1[C@@H](O)[C@@H](O)[C@@H](CO)O[C@H]1O[C@@H]1[C@@H](CO)O[C@H](O)[C@H](O)[C@H]1O GUBGYTABKSRVRQ-XLOQQCSPSA-N 0.000 description 1
- QGZKDVFQNNGYKY-UHFFFAOYSA-O Ammonium Chemical compound [NH4+] QGZKDVFQNNGYKY-UHFFFAOYSA-O 0.000 description 1
- 108010032595 Antibody Binding Sites Proteins 0.000 description 1
- 241000271566 Aves Species 0.000 description 1
- 102100032957 C5a anaphylatoxin chemotactic receptor 1 Human genes 0.000 description 1
- OYPRJOBELJOOCE-UHFFFAOYSA-N Calcium Chemical compound [Ca] OYPRJOBELJOOCE-UHFFFAOYSA-N 0.000 description 1
- 206010007559 Cardiac failure congestive Diseases 0.000 description 1
- 241001193938 Cavia magna Species 0.000 description 1
- 241000700199 Cavia porcellus Species 0.000 description 1
- 241000282693 Cercopithecidae Species 0.000 description 1
- 208000017667 Chronic Disease Diseases 0.000 description 1
- 102100022133 Complement C3 Human genes 0.000 description 1
- 108010028780 Complement C3 Proteins 0.000 description 1
- 102000016574 Complement C3-C5 Convertases Human genes 0.000 description 1
- 108010067641 Complement C3-C5 Convertases Proteins 0.000 description 1
- 108010034753 Complement Membrane Attack Complex Proteins 0.000 description 1
- 206010011224 Cough Diseases 0.000 description 1
- 208000011231 Crohn disease Diseases 0.000 description 1
- 206010050685 Cytokine storm Diseases 0.000 description 1
- 101710121417 Envelope glycoprotein Proteins 0.000 description 1
- 241000588724 Escherichia coli Species 0.000 description 1
- 241000206602 Eukaryota Species 0.000 description 1
- 108010008177 Fd immunoglobulins Proteins 0.000 description 1
- 108091006027 G proteins Proteins 0.000 description 1
- 102000003688 G-Protein-Coupled Receptors Human genes 0.000 description 1
- 108090000045 G-Protein-Coupled Receptors Proteins 0.000 description 1
- 102000030782 GTP binding Human genes 0.000 description 1
- 108091000058 GTP-Binding Proteins 0.000 description 1
- 108010010803 Gelatin Proteins 0.000 description 1
- 239000004471 Glycine Substances 0.000 description 1
- 206010019280 Heart failures Diseases 0.000 description 1
- 241000282412 Homo Species 0.000 description 1
- 101100440311 Homo sapiens C5 gene Proteins 0.000 description 1
- 101000867983 Homo sapiens C5a anaphylatoxin chemotactic receptor 1 Proteins 0.000 description 1
- 241000713772 Human immunodeficiency virus 1 Species 0.000 description 1
- 206010020751 Hypersensitivity Diseases 0.000 description 1
- 206010020772 Hypertension Diseases 0.000 description 1
- 206010021143 Hypoxia Diseases 0.000 description 1
- DGAQECJNVWCQMB-PUAWFVPOSA-M Ilexoside XXIX Chemical compound C[C@@H]1CC[C@@]2(CC[C@@]3(C(=CC[C@H]4[C@]3(CC[C@@H]5[C@@]4(CC[C@@H](C5(C)C)OS(=O)(=O)[O-])C)C)[C@@H]2[C@]1(C)O)C)C(=O)O[C@H]6[C@@H]([C@H]([C@@H]([C@H](O6)CO)O)O)O.[Na+] DGAQECJNVWCQMB-PUAWFVPOSA-M 0.000 description 1
- 206010022004 Influenza like illness Diseases 0.000 description 1
- 241001500351 Influenzavirus A Species 0.000 description 1
- 241001500350 Influenzavirus B Species 0.000 description 1
- 102100034349 Integrase Human genes 0.000 description 1
- 108090001007 Interleukin-8 Proteins 0.000 description 1
- 102000004890 Interleukin-8 Human genes 0.000 description 1
- 230000004163 JAK-STAT signaling pathway Effects 0.000 description 1
- ONIBWKKTOPOVIA-BYPYZUCNSA-N L-Proline Chemical compound OC(=O)[C@@H]1CCCN1 ONIBWKKTOPOVIA-BYPYZUCNSA-N 0.000 description 1
- HNDVDQJCIGZPNO-YFKPBYRVSA-N L-histidine Chemical compound OC(=O)[C@@H](N)CC1=CN=CN1 HNDVDQJCIGZPNO-YFKPBYRVSA-N 0.000 description 1
- GUBGYTABKSRVRQ-QKKXKWKRSA-N Lactose Natural products OC[C@H]1O[C@@H](O[C@H]2[C@H](O)[C@@H](O)C(O)O[C@@H]2CO)[C@H](O)[C@@H](O)[C@H]1O GUBGYTABKSRVRQ-QKKXKWKRSA-N 0.000 description 1
- 206010025323 Lymphomas Diseases 0.000 description 1
- 102000043136 MAP kinase family Human genes 0.000 description 1
- 108091054455 MAP kinase family Proteins 0.000 description 1
- 108010048043 Macrophage Migration-Inhibitory Factors Proteins 0.000 description 1
- 102000009073 Macrophage Migration-Inhibitory Factors Human genes 0.000 description 1
- 241000699670 Mus sp. Species 0.000 description 1
- GXCLVBGFBYZDAG-UHFFFAOYSA-N N-[2-(1H-indol-3-yl)ethyl]-N-methylprop-2-en-1-amine Chemical compound CN(CCC1=CNC2=C1C=CC=C2)CC=C GXCLVBGFBYZDAG-UHFFFAOYSA-N 0.000 description 1
- 206010028980 Neoplasm Diseases 0.000 description 1
- 208000036110 Neuroinflammatory disease Diseases 0.000 description 1
- 240000007594 Oryza sativa Species 0.000 description 1
- 235000007164 Oryza sativa Nutrition 0.000 description 1
- 241000282579 Pan Species 0.000 description 1
- 241000282577 Pan troglodytes Species 0.000 description 1
- 235000019483 Peanut oil Nutrition 0.000 description 1
- 108010079855 Peptide Aptamers Proteins 0.000 description 1
- 206010057249 Phagocytosis Diseases 0.000 description 1
- 206010035226 Plasma cell myeloma Diseases 0.000 description 1
- ZLMJMSJWJFRBEC-UHFFFAOYSA-N Potassium Chemical compound [K] ZLMJMSJWJFRBEC-UHFFFAOYSA-N 0.000 description 1
- 208000023146 Pre-existing disease Diseases 0.000 description 1
- 241000288906 Primates Species 0.000 description 1
- ONIBWKKTOPOVIA-UHFFFAOYSA-N Proline Natural products OC(=O)C1CCCN1 ONIBWKKTOPOVIA-UHFFFAOYSA-N 0.000 description 1
- 108010029485 Protein Isoforms Proteins 0.000 description 1
- 102000001708 Protein Isoforms Human genes 0.000 description 1
- 201000001263 Psoriatic Arthritis Diseases 0.000 description 1
- 208000036824 Psoriatic arthropathy Diseases 0.000 description 1
- 241000700159 Rattus Species 0.000 description 1
- 208000033626 Renal failure acute Diseases 0.000 description 1
- 241000725643 Respiratory syncytial virus Species 0.000 description 1
- 102100040756 Rhodopsin Human genes 0.000 description 1
- 108090000820 Rhodopsin Proteins 0.000 description 1
- 241000283984 Rodentia Species 0.000 description 1
- 240000004808 Saccharomyces cerevisiae Species 0.000 description 1
- 206010053879 Sepsis syndrome Diseases 0.000 description 1
- VYPSYNLAJGMNEJ-UHFFFAOYSA-N Silicium dioxide Chemical compound O=[Si]=O VYPSYNLAJGMNEJ-UHFFFAOYSA-N 0.000 description 1
- 229920002472 Starch Polymers 0.000 description 1
- CZMRCDWAGMRECN-UGDNZRGBSA-N Sucrose Chemical compound O[C@H]1[C@H](O)[C@@H](CO)O[C@@]1(CO)O[C@@H]1[C@H](O)[C@@H](O)[C@H](O)[C@@H](CO)O1 CZMRCDWAGMRECN-UGDNZRGBSA-N 0.000 description 1
- 229930006000 Sucrose Natural products 0.000 description 1
- 206010051379 Systemic Inflammatory Response Syndrome Diseases 0.000 description 1
- FEWJPZIEWOKRBE-UHFFFAOYSA-N Tartaric Acid Chemical class [H+].[H+].[O-]C(=O)C(O)C(O)C([O-])=O FEWJPZIEWOKRBE-UHFFFAOYSA-N 0.000 description 1
- 238000002835 absorbance Methods 0.000 description 1
- 230000002378 acidificating effect Effects 0.000 description 1
- 239000013543 active substance Substances 0.000 description 1
- 230000001154 acute effect Effects 0.000 description 1
- 201000011040 acute kidney failure Diseases 0.000 description 1
- 230000004658 acute-phase response Effects 0.000 description 1
- 230000003044 adaptive effect Effects 0.000 description 1
- 230000000996 additive effect Effects 0.000 description 1
- 210000001789 adipocyte Anatomy 0.000 description 1
- 239000002671 adjuvant Substances 0.000 description 1
- 125000003545 alkoxy group Chemical group 0.000 description 1
- 125000002947 alkylene group Chemical group 0.000 description 1
- 208000026935 allergic disease Diseases 0.000 description 1
- 210000002821 alveolar epithelial cell Anatomy 0.000 description 1
- 125000003277 amino group Chemical group 0.000 description 1
- 238000013103 analytical ultracentrifugation Methods 0.000 description 1
- 230000003466 anti-cipated effect Effects 0.000 description 1
- 238000011319 anticancer therapy Methods 0.000 description 1
- 239000003146 anticoagulant agent Substances 0.000 description 1
- 229940127219 anticoagulant drug Drugs 0.000 description 1
- 230000010100 anticoagulation Effects 0.000 description 1
- 239000000607 artificial tear Substances 0.000 description 1
- PUKBOVABABRILL-YZNIXAGQSA-N avacopan Chemical group C1=C(C(F)(F)F)C(C)=CC=C1NC(=O)[C@@H]1[C@H](C=2C=CC(NC3CCCC3)=CC=2)N(C(=O)C=2C(=CC=CC=2C)F)CCC1 PUKBOVABABRILL-YZNIXAGQSA-N 0.000 description 1
- 229950001740 avacopan Drugs 0.000 description 1
- 210000003651 basophil Anatomy 0.000 description 1
- 230000009286 beneficial effect Effects 0.000 description 1
- 239000011230 binding agent Substances 0.000 description 1
- 230000003115 biocidal effect Effects 0.000 description 1
- 210000000601 blood cell Anatomy 0.000 description 1
- 238000004820 blood count Methods 0.000 description 1
- 210000001772 blood platelet Anatomy 0.000 description 1
- 210000001601 blood-air barrier Anatomy 0.000 description 1
- 210000001185 bone marrow Anatomy 0.000 description 1
- 238000010322 bone marrow transplantation Methods 0.000 description 1
- 210000004899 c-terminal region Anatomy 0.000 description 1
- 239000011575 calcium Substances 0.000 description 1
- 229910052791 calcium Inorganic materials 0.000 description 1
- 239000002775 capsule Substances 0.000 description 1
- 125000002837 carbocyclic group Chemical group 0.000 description 1
- 125000004432 carbon atom Chemical group C* 0.000 description 1
- 230000002612 cardiopulmonary effect Effects 0.000 description 1
- 230000001364 causal effect Effects 0.000 description 1
- 239000003153 chemical reaction reagent Substances 0.000 description 1
- 230000001684 chronic effect Effects 0.000 description 1
- 238000003776 cleavage reaction Methods 0.000 description 1
- 230000015271 coagulation Effects 0.000 description 1
- 238000005345 coagulation Methods 0.000 description 1
- 238000002742 combinatorial mutagenesis Methods 0.000 description 1
- 230000009850 completed effect Effects 0.000 description 1
- 230000001010 compromised effect Effects 0.000 description 1
- 238000012790 confirmation Methods 0.000 description 1
- 238000007796 conventional method Methods 0.000 description 1
- 208000029078 coronary artery disease Diseases 0.000 description 1
- 239000003246 corticosteroid Substances 0.000 description 1
- 229960001334 corticosteroids Drugs 0.000 description 1
- 229940109239 creatinine Drugs 0.000 description 1
- 230000009260 cross reactivity Effects 0.000 description 1
- 239000013078 crystal Substances 0.000 description 1
- 206010052015 cytokine release syndrome Diseases 0.000 description 1
- 230000000254 damaging effect Effects 0.000 description 1
- 230000002498 deadly effect Effects 0.000 description 1
- 230000007123 defense Effects 0.000 description 1
- 238000012217 deletion Methods 0.000 description 1
- 230000037430 deletion Effects 0.000 description 1
- 238000000586 desensitisation Methods 0.000 description 1
- 229960003957 dexamethasone Drugs 0.000 description 1
- UREBDLICKHMUKA-CXSFZGCWSA-N dexamethasone Chemical compound C1CC2=CC(=O)C=C[C@]2(C)[C@]2(F)[C@@H]1[C@@H]1C[C@@H](C)[C@@](C(=O)CO)(O)[C@@]1(C)C[C@@H]2O UREBDLICKHMUKA-CXSFZGCWSA-N 0.000 description 1
- 239000008121 dextrose Substances 0.000 description 1
- 206010012601 diabetes mellitus Diseases 0.000 description 1
- 238000000502 dialysis Methods 0.000 description 1
- 230000004069 differentiation Effects 0.000 description 1
- 239000003085 diluting agent Substances 0.000 description 1
- 231100000673 dose–response relationship Toxicity 0.000 description 1
- 230000007783 downstream signaling Effects 0.000 description 1
- 208000017574 dry cough Diseases 0.000 description 1
- 230000009977 dual effect Effects 0.000 description 1
- 230000004064 dysfunction Effects 0.000 description 1
- 230000002526 effect on cardiovascular system Effects 0.000 description 1
- 239000012636 effector Substances 0.000 description 1
- 238000002565 electrocardiography Methods 0.000 description 1
- 239000003792 electrolyte Substances 0.000 description 1
- 239000008144 emollient laxative Substances 0.000 description 1
- 239000003995 emulsifying agent Substances 0.000 description 1
- 239000000839 emulsion Substances 0.000 description 1
- 210000003979 eosinophil Anatomy 0.000 description 1
- 210000003743 erythrocyte Anatomy 0.000 description 1
- 238000011156 evaluation Methods 0.000 description 1
- 235000013861 fat-free Nutrition 0.000 description 1
- 235000013312 flour Nutrition 0.000 description 1
- 238000000684 flow cytometry Methods 0.000 description 1
- 239000008273 gelatin Substances 0.000 description 1
- 229920000159 gelatin Polymers 0.000 description 1
- 235000019322 gelatine Nutrition 0.000 description 1
- 235000011852 gelatine desserts Nutrition 0.000 description 1
- 230000002068 genetic effect Effects 0.000 description 1
- 102000034238 globular proteins Human genes 0.000 description 1
- 108091005896 globular proteins Proteins 0.000 description 1
- 239000008103 glucose Substances 0.000 description 1
- YQEMORVAKMFKLG-UHFFFAOYSA-N glycerine monostearate Natural products CCCCCCCCCCCCCCCCCC(=O)OC(CO)CO YQEMORVAKMFKLG-UHFFFAOYSA-N 0.000 description 1
- SVUQHVRAGMNPLW-UHFFFAOYSA-N glycerol monostearate Natural products CCCCCCCCCCCCCCCCC(=O)OCC(O)CO SVUQHVRAGMNPLW-UHFFFAOYSA-N 0.000 description 1
- 125000003712 glycosamine group Chemical group 0.000 description 1
- 230000012010 growth Effects 0.000 description 1
- 239000001963 growth medium Substances 0.000 description 1
- 230000003394 haemopoietic effect Effects 0.000 description 1
- 125000001188 haloalkyl group Chemical group 0.000 description 1
- 210000002216 heart Anatomy 0.000 description 1
- 210000003958 hematopoietic stem cell Anatomy 0.000 description 1
- 210000003494 hepatocyte Anatomy 0.000 description 1
- 125000000623 heterocyclic group Chemical group 0.000 description 1
- HNDVDQJCIGZPNO-UHFFFAOYSA-N histidine Natural products OC(=O)C(N)CC1=CN=CN1 HNDVDQJCIGZPNO-UHFFFAOYSA-N 0.000 description 1
- 230000013632 homeostatic process Effects 0.000 description 1
- 210000005260 human cell Anatomy 0.000 description 1
- 238000011577 humanized mouse model Methods 0.000 description 1
- 150000004677 hydrates Chemical class 0.000 description 1
- 150000004679 hydroxides Chemical class 0.000 description 1
- 125000002887 hydroxy group Chemical group [H]O* 0.000 description 1
- WGCNASOHLSPBMP-UHFFFAOYSA-N hydroxyacetaldehyde Natural products OCC=O WGCNASOHLSPBMP-UHFFFAOYSA-N 0.000 description 1
- 230000009610 hypersensitivity Effects 0.000 description 1
- 230000036044 hypoxaemia Effects 0.000 description 1
- 125000002883 imidazolyl group Chemical group 0.000 description 1
- 230000003832 immune regulation Effects 0.000 description 1
- 208000026278 immune system disease Diseases 0.000 description 1
- 230000002163 immunogen Effects 0.000 description 1
- 239000002955 immunomodulating agent Substances 0.000 description 1
- 229940121354 immunomodulator Drugs 0.000 description 1
- 230000002584 immunomodulator Effects 0.000 description 1
- 238000000338 in vitro Methods 0.000 description 1
- 238000001727 in vivo Methods 0.000 description 1
- 230000008595 infiltration Effects 0.000 description 1
- 238000001764 infiltration Methods 0.000 description 1
- 230000002757 inflammatory effect Effects 0.000 description 1
- 239000004615 ingredient Substances 0.000 description 1
- 229940125369 inhaled corticosteroids Drugs 0.000 description 1
- 230000000977 initiatory effect Effects 0.000 description 1
- 238000003780 insertion Methods 0.000 description 1
- 230000037431 insertion Effects 0.000 description 1
- 230000003434 inspiratory effect Effects 0.000 description 1
- 230000017306 interleukin-6 production Effects 0.000 description 1
- 230000031146 intracellular signal transduction Effects 0.000 description 1
- 230000004068 intracellular signaling Effects 0.000 description 1
- JJWLVOIRVHMVIS-UHFFFAOYSA-N isopropylamine Chemical compound CC(C)N JJWLVOIRVHMVIS-UHFFFAOYSA-N 0.000 description 1
- 238000011813 knockout mouse model Methods 0.000 description 1
- 239000008101 lactose Substances 0.000 description 1
- 239000008141 laxative Substances 0.000 description 1
- 229940125722 laxative agent Drugs 0.000 description 1
- 208000019423 liver disease Diseases 0.000 description 1
- 238000011068 loading method Methods 0.000 description 1
- 229940125386 long-acting bronchodilator Drugs 0.000 description 1
- 210000004698 lymphocyte Anatomy 0.000 description 1
- 230000000527 lymphocytic effect Effects 0.000 description 1
- 238000011418 maintenance treatment Methods 0.000 description 1
- 230000003211 malignant effect Effects 0.000 description 1
- 239000000463 material Substances 0.000 description 1
- 235000013336 milk Nutrition 0.000 description 1
- 239000008267 milk Substances 0.000 description 1
- 210000004080 milk Anatomy 0.000 description 1
- 239000002480 mineral oil Substances 0.000 description 1
- 235000010446 mineral oil Nutrition 0.000 description 1
- 238000012544 monitoring process Methods 0.000 description 1
- 125000002757 morpholinyl group Chemical group 0.000 description 1
- 230000008383 multiple organ dysfunction Effects 0.000 description 1
- 230000035772 mutation Effects 0.000 description 1
- 201000000050 myeloid neoplasm Diseases 0.000 description 1
- 230000003959 neuroinflammation Effects 0.000 description 1
- 230000007658 neurological function Effects 0.000 description 1
- 210000002569 neuron Anatomy 0.000 description 1
- 230000004112 neuroprotection Effects 0.000 description 1
- 238000010606 normalization Methods 0.000 description 1
- 150000007523 nucleic acids Chemical group 0.000 description 1
- 235000016709 nutrition Nutrition 0.000 description 1
- 239000003921 oil Substances 0.000 description 1
- 235000019198 oils Nutrition 0.000 description 1
- 239000002674 ointment Substances 0.000 description 1
- 229940124624 oral corticosteroid Drugs 0.000 description 1
- 230000008816 organ damage Effects 0.000 description 1
- 239000006179 pH buffering agent Substances 0.000 description 1
- UOJMTSCORVQOHS-UHFFFAOYSA-N pachypodol Natural products COc1cc(ccc1O)C2=C(C)C(=O)c3c(O)cc(C)cc3O2 UOJMTSCORVQOHS-UHFFFAOYSA-N 0.000 description 1
- 230000036961 partial effect Effects 0.000 description 1
- 244000052769 pathogen Species 0.000 description 1
- 239000000312 peanut oil Substances 0.000 description 1
- 230000035699 permeability Effects 0.000 description 1
- 102000013415 peroxidase activity proteins Human genes 0.000 description 1
- 108040007629 peroxidase activity proteins Proteins 0.000 description 1
- 239000003208 petroleum Substances 0.000 description 1
- 230000008782 phagocytosis Effects 0.000 description 1
- 239000008194 pharmaceutical composition Substances 0.000 description 1
- 229940124531 pharmaceutical excipient Drugs 0.000 description 1
- 239000006187 pill Substances 0.000 description 1
- 125000003386 piperidinyl group Chemical group 0.000 description 1
- 108091033319 polynucleotide Proteins 0.000 description 1
- 102000040430 polynucleotide Human genes 0.000 description 1
- 239000002157 polynucleotide Substances 0.000 description 1
- 239000011148 porous material Substances 0.000 description 1
- 239000011591 potassium Substances 0.000 description 1
- 229910052700 potassium Inorganic materials 0.000 description 1
- 230000003389 potentiating effect Effects 0.000 description 1
- 239000000843 powder Substances 0.000 description 1
- 230000002265 prevention Effects 0.000 description 1
- 230000007112 pro inflammatory response Effects 0.000 description 1
- MFDFERRIHVXMIY-UHFFFAOYSA-N procaine Chemical compound CCN(CC)CCOC(=O)C1=CC=C(N)C=C1 MFDFERRIHVXMIY-UHFFFAOYSA-N 0.000 description 1
- 229960004919 procaine Drugs 0.000 description 1
- 230000000770 proinflammatory effect Effects 0.000 description 1
- QQONPFPTGQHPMA-UHFFFAOYSA-N propylene Natural products CC=C QQONPFPTGQHPMA-UHFFFAOYSA-N 0.000 description 1
- 125000004805 propylene group Chemical group [H]C([H])([H])C([H])([*:1])C([H])([H])[*:2] 0.000 description 1
- 230000001681 protective effect Effects 0.000 description 1
- 230000006337 proteolytic cleavage Effects 0.000 description 1
- 125000000714 pyrimidinyl group Chemical group 0.000 description 1
- 125000001422 pyrrolinyl group Chemical group 0.000 description 1
- 125000000168 pyrrolyl group Chemical group 0.000 description 1
- 238000002708 random mutagenesis Methods 0.000 description 1
- 239000003642 reactive oxygen metabolite Substances 0.000 description 1
- 238000010188 recombinant method Methods 0.000 description 1
- 230000007115 recruitment Effects 0.000 description 1
- 230000029964 regulation of glucose metabolic process Effects 0.000 description 1
- 230000028503 regulation of lipid metabolic process Effects 0.000 description 1
- 230000019254 respiratory burst Effects 0.000 description 1
- 210000002345 respiratory system Anatomy 0.000 description 1
- 235000009566 rice Nutrition 0.000 description 1
- 238000011808 rodent model Methods 0.000 description 1
- 229920006395 saturated elastomer Polymers 0.000 description 1
- 230000007017 scission Effects 0.000 description 1
- 210000002966 serum Anatomy 0.000 description 1
- 239000008159 sesame oil Substances 0.000 description 1
- 235000011803 sesame oil Nutrition 0.000 description 1
- 239000000741 silica gel Substances 0.000 description 1
- 229910002027 silica gel Inorganic materials 0.000 description 1
- 238000002741 site-directed mutagenesis Methods 0.000 description 1
- 235000020183 skimmed milk Nutrition 0.000 description 1
- 239000011734 sodium Substances 0.000 description 1
- 229910052708 sodium Inorganic materials 0.000 description 1
- RYYKJJJTJZKILX-UHFFFAOYSA-M sodium octadecanoate Chemical compound [Na+].CCCCCCCCCCCCCCCCCC([O-])=O RYYKJJJTJZKILX-UHFFFAOYSA-M 0.000 description 1
- 239000002904 solvent Substances 0.000 description 1
- 210000001082 somatic cell Anatomy 0.000 description 1
- 230000037439 somatic mutation Effects 0.000 description 1
- 238000001179 sorption measurement Methods 0.000 description 1
- 239000003549 soybean oil Substances 0.000 description 1
- 235000012424 soybean oil Nutrition 0.000 description 1
- 239000008107 starch Substances 0.000 description 1
- 235000019698 starch Nutrition 0.000 description 1
- 238000000551 statistical hypothesis test Methods 0.000 description 1
- 150000003431 steroids Chemical class 0.000 description 1
- 230000004936 stimulating effect Effects 0.000 description 1
- 239000005720 sucrose Substances 0.000 description 1
- 230000008093 supporting effect Effects 0.000 description 1
- 238000009120 supportive therapy Methods 0.000 description 1
- 239000000829 suppository Substances 0.000 description 1
- 239000000725 suspension Substances 0.000 description 1
- 238000013268 sustained release Methods 0.000 description 1
- 239000012730 sustained-release form Substances 0.000 description 1
- 229940053017 sylvant Drugs 0.000 description 1
- 238000003786 synthesis reaction Methods 0.000 description 1
- 230000009885 systemic effect Effects 0.000 description 1
- 201000000596 systemic lupus erythematosus Diseases 0.000 description 1
- 239000003826 tablet Substances 0.000 description 1
- 239000000454 talc Substances 0.000 description 1
- 229910052623 talc Inorganic materials 0.000 description 1
- 230000008685 targeting Effects 0.000 description 1
- 235000002906 tartaric acid Nutrition 0.000 description 1
- 125000003718 tetrahydrofuranyl group Chemical group 0.000 description 1
- 125000001412 tetrahydropyranyl group Chemical group 0.000 description 1
- 230000001225 therapeutic effect Effects 0.000 description 1
- 231100000827 tissue damage Toxicity 0.000 description 1
- 230000017423 tissue regeneration Effects 0.000 description 1
- 231100000419 toxicity Toxicity 0.000 description 1
- 230000001988 toxicity Effects 0.000 description 1
- 150000003626 triacylglycerols Chemical class 0.000 description 1
- 125000002023 trifluoromethyl group Chemical group FC(F)(F)* 0.000 description 1
- 230000003827 upregulation Effects 0.000 description 1
- 210000002700 urine Anatomy 0.000 description 1
- 238000005353 urine analysis Methods 0.000 description 1
- 238000002255 vaccination Methods 0.000 description 1
- 229940124549 vasodilator Drugs 0.000 description 1
- 239000003071 vasodilator agent Substances 0.000 description 1
- 235000013311 vegetables Nutrition 0.000 description 1
- 239000003981 vehicle Substances 0.000 description 1
- 238000009423 ventilation Methods 0.000 description 1
- 230000002861 ventricular Effects 0.000 description 1
- 230000029812 viral genome replication Effects 0.000 description 1
- 230000009385 viral infection Effects 0.000 description 1
- 230000000007 visual effect Effects 0.000 description 1
- 239000011782 vitamin Substances 0.000 description 1
- 235000013343 vitamin Nutrition 0.000 description 1
- 229940088594 vitamin Drugs 0.000 description 1
- 229930003231 vitamin Natural products 0.000 description 1
- 230000003442 weekly effect Effects 0.000 description 1
- 238000009736 wetting Methods 0.000 description 1
- 239000000080 wetting agent Substances 0.000 description 1
- 210000005253 yeast cell Anatomy 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K45/00—Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
- A61K45/06—Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
-
- 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
- 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
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/28—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
- C07K16/2866—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for cytokines, lymphokines, interferons
-
- 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
-
- 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
- A61K2039/507—Comprising a combination of two or more separate antibodies
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/20—Immunoglobulins specific features characterized by taxonomic origin
- C07K2317/24—Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
Definitions
- the present invention relates to an inhibitor of C5a activity and an inhibitor of IL-6 activity for use in the treatment of infectious pneumonia and infectious acute respiratory distress syndrome (ARDS).
- ARDS infectious acute respiratory distress syndrome
- administration regimes and kit of parts are referred to.
- C5a is one of the most potent inflammatory peptides, with a broad spectrum of functions (Guo and Ward, 2005). C5a exerts its effects through the high-affinity C5a receptors (C5aR and C5L2) (Ward, 2009). C5aR belongs to the rhodopsin family of G-protein-coupled receptors with seven transmembrane segments; C5L2 has a similar structure but appears not to be G-protein-coupled. It is currently believed that C5a exerts its biological functions primarily through C5a-C5aR interaction. However, some reports demonstrate signaling also through C5L2 activation (Rittirsch and others, 2008).
- C5aR is widely expressed on myeloid cells including neutrophils, eosinophils, basophils, and monocytes, and non-myeloid cells in many organs, especially in the lung and liver, indicative of the importance of C5a/C5aR signaling.
- Widespread up-regulation of C5aR expression occurs during the onset of sepsis, and blockade of C5a/C5aR interaction by anti- C5a, or anti-C5aR antibodies, or C5aR antagonists renders highly protective effects in rodent models of sepsis (Czermak and others, 1999; Huber-Lang and others, 2001; Riedemann and others, 2002).
- C5a has been reported to exert numerous pro-inflammatory responses.
- C5a stimulates the synthesis and release from human leukocytes of pro-inflammatory cytokines such as TNF-a, IL-ip, IL-6, IL-8, and macrophage migration inhibitory factor (MIF) (Hopken U et al. 1996. Eur J Immunol 26(5): 1103-1109; Riedemann NC et al. 2004. J Immunol 173(2): 1355-1359; Stricter RM et al. 1992. Am J Pathol 141(2):397-407).
- pro-inflammatory cytokines such as TNF-a, IL-ip, IL-6, IL-8, and macrophage migration inhibitory factor (MIF)
- C5a produces a strong synergistic effect with LPS in production of TNF-a, macrophage inflammatory protein (MIP)-2, cytokine-induced neutrophil chemoattractant (CINC)-l, and IL-ip in alveolar epithelial cells (Riedemann NC et al. 2002. J. Immunol. 168(4): 1919-1925; Rittirsch D et al. 2008. Nat Rev Immunol 8(10):776-787).
- MIP macrophage inflammatory protein
- CINC cytokine-induced neutrophil chemoattractant
- C5a has a variety of biological functions (Guo and Ward, 2005).
- C5a is a strong chemoattractant for neutrophils and also has chemotactic activity for monocytes and macrophages.
- C5a causes an oxidative burst (02 consumption) in neutrophils and enhances phagocytosis and release of granular enzymes.
- C5a has also been found to be a vasodilator.
- C5a has been shown to be involved in modulation of cytokine expression from various cell types and to enhance expression of adhesion molecule expression on neutrophils. High doses of C5a can lead to nonspecific chemotactic “desensitization” of neutrophils, thereby causing broad dysfunction.
- Interleukin-6 IL-6
- IL-6 has a fundamental role in inflammation, immune regulation, hematopoiesis, host defense, homeostasis, and tissue regeneration. It is produced by a wide range of hematopoietic and somatic cells that influences numerous cell types with multiple biological functions.
- IL-6 plays a critical role in the immune response by stimulating the growth and differentiation of T and B cells, as well as promoting the production of antibodies. Further, IL- 6 is involved in the acute phase response to injury or infection, promoting inflammation and activating immune cells. Its function in hematopoiesis is to stimulate the production of blood cells in the bone marrow, including red blood cells, white blood cells, and platelets. IL-6 is further involved in the regulation of glucose metabolism and lipid metabolism and has been implicated in various neurological functions, including neuroprotection and neuroinflammation. Furthermore, abnormal IL-6 production has been associated with the development of a wide variety of systemic immune-mediated, chronic diseases, and even neoplasms.
- IL-6 transmits its signals through a cell-surface type-I receptor complex, that consists of a ligand-binding glycoprotein termed as IL-6 receptor (IL-6R) and a signal-transducing component gpl30.
- IL-6 receptor IL-6 receptor
- gpl30 a signal-transducing component
- IL-6R ligand-binding glycoprotein
- gpl30 signal-transducing component
- IL-6 receptor binds to IL-6R, it induces a conformational change that enables the recruitment and assembly of gp!30. This leads to the formation of a high-affinity IL-6 receptor complex, which activates intracellular signaling pathways.
- the downstream signaling pathways activated by the IL-6 receptor complex depend on the cell type and context but can include the JAK-STAT pathway, the PI3K-Akt pathway, and the MAPK pathway.
- IL-6R Two forms of IL-6R are known: a membrane-bound form (mIL-6R) and a soluble form (sIL-6R).
- mIL-6R membrane-bound form
- sIL-6R soluble form
- the membrane-bound form is found on the surface of cells and is responsible for the high-affinity binding of IL-6.
- the soluble form is generated by proteolytic cleavage of mIL-6R and can bind to IL-6, forming a complex with gpl30 and inducing intracellular signaling.
- the sIL-6R is capable of activating cells that do not express the membrane-bound form of IL-6R, expanding the range of cells that can respond to IL-6.
- the IL-6 receptor is expressed on a wide range of cells, including immune cells, hepatocytes, adipocytes, and neurons.
- SARS Severe Acute Respiratory Syndrome
- MERS Middle East Respiratory Syndrome
- SARS-CoV-2 Coronavirus disease 2019
- COVID-19 typically presents with features of long non-symptomatic latency which might be a main contributing factor to a relatively high transmissibility compared to the other previously occurring deadly coronavirus infections, severe acute respiratory syndrome (SARS) and Middle East respiratory syndrome (MERS).
- COVID-19 patients typically present with flulike symptoms such as fever or signs of lower respiratory tract illness including dry cough and shortness of. With the progression of disease into a severe form, it often affects multiple organs’ functions including the lung, heart, liver, and coagulation system among others. As such, death is typically caused by respiratory failure and multiple organ dysfunctions similar to other viral pneumonia-induced sepsis. Sepsis and ARDS mostly occur in the second week upon disease onset.
- COVID-19 can be characterized by a dual play of viral inflammation and immune- mediated injury that may result in a “complement storm” event occurring in the progression of corona virus infection, in particular COVID-19.
- Anti-C5a therapy for COVID-19 has been disclosed in W02021/190770 Al and recently received emergency use authorization (EUA) by the FDA.
- EUA emergency use authorization
- Tocilizumab also known as Actemra
- EUA and BLA from the FDA.
- the surprising effect of a combined anti-C5a and anti-IL-6 therapy of infectious pneumonia and/or infectious acute respiratory distress syndrome (ARDS) is disclosed.
- the present invention provides an inhibitor of C5a activity for use in:
- the present invention provides an inhibitor of IL-6 activity for use in:
- the present invention provides a kit of parts comprising an inhibitor of C5a activity and an inhibitor of IL-6 activity.
- FIG. 1 IMP Administrations in Phase III refers to the administration schedule of the investigational medicinal product (IMP).
- the anti-C5a antibody IFX-1 (vilobelimab) was administered at a dose of 800mg on several days during the treatment period as indicated in figure 1.
- Figure 2 60-day all-cause mortality for treatment with vilobelimab and any use of tocilizumab.
- Line 1 Vilo + SOC with any use of tocilizumab
- Line 2 Vilo + SOC without any use of tocilizumab
- Line 3 Placebo + SOC with any use of tocvilizumab
- Line 4 Placebo + SOC without any use of tocilizumab;
- Figure 3 60-day all-cause mortality for treatment with vilobelimab and baseline use of tocilizumab.
- Line 1 Vilo + SOC with baseline use of tocilizumab
- Line 2 Vilo + SOC without baseline use of tocilizumab
- Line 3 Placebo + SOC with any use of tocilizumab
- Line 4 Placebo + SOC without baseline use of tocilizumab;
- the terms used herein are defined as described in "A multilingual glossary of biotechnological terms: (IUPAC Recommendations)", Leuenberger, H.G.W, Nagel, B. and Klbl, H. eds. (1995), Helvetica Chimica Acta, CH-4010 Basel, Switzerland).
- C5a particularly refers to human C5a.
- Human C5a is a 74 amino acid peptide with the following amino acid sequence:
- TLQKKIEEIA AKYKHSWKK CCYDGACVNN DETCEQRAAR ISLGPRCIKA FTECCWASQ LRANISHKDM QLGR (SEQ ID NO: 1).
- inhibitor of C5a activity refers to any compound that in any way reduces the activity of C5a. This activity reduction can be achieved by directly or indirectly lowering the concentration of C5a, or by reducing the activity of C5a, or by preventing that C5a exerts its effects on one or more of its receptors (e.g. on C5aR or C5L2), or by reducing the concentration or activity of one or more receptors of C5a.
- Preferred inhibitors of C5a activity are antibodies specifically binding to hC5a.
- C5a receptor refers to any potential C5a binding ligand on the cell surface, especially to any receptor protein to which C5a may bind and elicit a reaction on said receptor (e.g. activation or inhibition of the receptor).
- C5a receptor particularly encompasses the two receptors C5aR and C5L2.
- Alternative names for C5aR are C5aRl and CD88.
- An alternative name for C5L2 is C5aR2.
- Interleukin-6 particularly refers to human IL-6.
- Human IL-6 is an 212 amino acid peptide.
- the amino acid sequence of human IL-6 can be found under the accession number UniProtKB P05231 (IL6 HUMAN).
- inhibitor of IL-6 activity refers to any compound that in any way reduces the activity of IL-6. This activity reduction can be achieved by directly or indirectly lowering the concentration of IL-6, or by reducing the activity of IL-6, or by preventing that IL-6 exerts its effects on one or more of its receptors (e.g. on IL-6R), or by reducing the concentration or activity of one or more receptors of IL-6.
- Preferred inhibitors of IL-6 activity are antibodies specifically binding to IL-6 or IL-6R, preferably IL-6R.
- IL-6 receptor refers to any potential IL-6 binding ligand, especially to any receptor protein to which IL-6 may bind and elicit a reaction on said receptor (e.g. activation or inhibition of the receptor).
- IL-6 receptor (IL-6R; sometimes referred to as CD 126) particularly encompasses the two receptor forms sIL-6R and mIL-6R.
- a first compound e.g. an antibody or antigen-binding fragment thereof
- a second compound e.g. a target protein
- a dissociation constant Ka to said second compound of 1 mM or less, preferably 100 pM or less, preferably 50 pM or less, preferably 30 pM or less, preferably 20 pM or less, preferably 10 pM or less, preferably 5 pM or less, more preferably 1 pM or less, more preferably 900 nM or less, more preferably 800 nM or less, more preferably 700 nM or less, more preferably 600 nM or less, more preferably 500 nM or less, more preferably 400 nM or less, more preferably 300 nM or less, more preferably 200 nM or less, even more preferably 100 nM or less, even more preferably 90 nM or less, even more preferably 80 nM or less, even more preferably 70 nM
- binding preferably relates to a specific binding.
- Specific binding means that a compound (e.g an antibody or antigen-binding fragment thereof) binds stronger to a target such as an epitope for which it is specific compared to the binding to another target.
- a compound binds stronger to a first target compared to a second target, if it binds to the first target with a dissociation constant (Ka) which is lower than the dissociation constant for the second target.
- Ka dissociation constant
- the dissociation constant (Ka) for the target to which the compound binds specifically is more than 10-fold, preferably more than 20- fold, more preferably more than 50-fold, even more preferably more than 100-fold, 200-fold, 500-fold or 1000-fold lower than the dissociation constant (Ka) for the target to which the compound does not bind specifically.
- Ka (usually measured in “mol/L”, sometimes abbreviated as “M”) is intended to refer to the dissociation equilibrium constant of the particular interaction between a binding moiety (e.g. an antibody or antigen-binding fragment thereof) and a target molecule (e.g. an antigen or epitope thereof).
- Methods for determining binding affinities of compounds are known to a person of ordinary skill in the art and can be selected for instance from the following methods known in the art: Surface Plasmon Resonance (SPR) based technology, Bio-layer interferometry (BLI), enzyme-linked immunosorbent assay (ELISA), flow cytometry, isothermal titration calorimetry (ITC), analytical ultracentrifugation, radioimmunoassay (RIA or IRMA) and enhanced chemiluminescence (ECL).
- SPR Surface Plasmon Resonance
- BBI Bio-layer interferometry
- ELISA enzyme-linked immunosorbent assay
- ITC isothermal titration calorimetry
- analytical ultracentrifugation RIA or IRMA
- ECL enhanced chemiluminescence
- the dissociation constant Ka is determined at 20°C, 25°C, 30°C, or 37°C. If not specifically indicated otherwise, the Ka values recited herein are determined by surface
- antibodies and antigen-binding fragments thereof according to the invention bind with a sufficient binding affinity to their target, for example, with a Kd value of between 500 nM-1 pM, i.e. 500 nM, 450 nM, 400nM, 350 nM, 300nM, 250 nM, 200nM, 150 nM, lOOnM, 50 nM, 10 nM, 1 nM, 900 pM, 800 pM, 700 pM, 600 pM, 500 pM, 400 pM, 300 pM, 200 pM, 100 pM, 50 pM, IpM.
- a Kd value of between 500 nM-1 pM, i.e. 500 nM, 450 nM, 400nM, 350 nM, 300nM, 250 nM, 200nM, 150 nM, lOOnM, 50 nM, 10 nM, 1 nM, 900 pM, 800
- an “epitope”, also known as antigenic determinant, is the part of a macromolecule that is recognized by the immune system, specifically by antibodies, B cells, or T cells.
- an “epitope” is the part of a macromolecule capable of binding to a compound (e.g. an antibody or antigen-binding fragment thereof) as described herein.
- binding preferably relates to a specific binding.
- Epitopes usually consist of chemically active surface groupings of molecules such as amino acids or sugar side chains and usually have specific three-dimensional structural characteristics, as well as specific charge characteristics. Conformational and non-conformational epitopes can be distinguished in that the binding to the former but not the latter is lost in the presence of denaturing solvents.
- a “paratope” is the part of an antibody that binds to the epitope.
- antibody typically refers to a glycoprotein comprising at least two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds, or an antigen-binding portion thereof.
- antibody also includes all recombinant forms of antibodies, in particular of the antibodies described herein, e.g. antibodies expressed in prokaryotes, unglycosylated antibodies, antibodies expressed in eukaryotes (e.g. CHO cells), glycosylated antibodies, and any antigen-binding antibody fragments and derivatives as described below.
- Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as VH or VH) and a heavy chain constant region.
- Each light chain is comprised of a light chain variable region (abbreviated herein as VL or VL) and a light chain constant region.
- VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR).
- CDR complementarity determining regions
- FR framework regions
- Each VH and VL is composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4.
- the variable regions of the heavy and light chains contain a binding domain that interacts with an antigen.
- the constant regions of the antibodies may mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the first component (Clq) of the classical complement system.
- CDR or “complementarity determining region” refers to the non-contiguous antigen binding sites found within the variable region of both heavy and light chain polypeptides (sometimes referred to as LCDR or HCDR).
- CDRs have been described by Kabat et al., J. Biol. Chem. 252:6609-6616 (1977); Kabat et al., U.S. Dept, of Health and Human Services, "Sequences of proteins of immunological interest” (1991) (also referred to herein as Kabat 1991); by Chothia et al., J. Mol. Biol. 196:901-917 (1987) (also referred to herein as Chothia 1987); and MacCallum et al., J.
- antigen-binding fragment of an antibody (or simply “binding portion”), as used herein, refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen. It has been shown that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody.
- binding fragments encompassed within the term “antigen-binding portion” of an antibody include (i) Fab fragments, monovalent fragments consisting of the VL, VH, CL and CH domains; (ii) F(ab')2 fragments, bivalent fragments comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) Fd fragments consisting of the VH and CH domains; (iv) Fv fragments consisting of the VL and VH domains of a single arm of an antibody, (v) dAb fragments (Ward et al., (1989) Nature 341 : 544-546), which consist of a VH domain; (vi) isolated complementarity determining regions (CDR), and (vii) combinations of two or more isolated CDRs which may optionally be joined by a synthetic linker.
- Fab fragments monovalent fragments consisting of the VL, VH, CL and CH domains
- F(ab')2 fragments bivalent fragments
- the two domains of the Fv fragment, VL and VH are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv); see e.g., Bird et al. (1988) Science 242: 423-426; and Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85: 5879- 5883).
- single chain Fv single chain Fv
- Such single chain antibodies are also intended to be encompassed within the term “antigen-binding fragment” of an antibody.
- a further example is a binding-domain immunoglobulin fusion protein comprising (i) a binding domain polypeptide that is fused to an immunoglobulin hinge region polypeptide, (ii) an immunoglobulin heavy chain CH2 constant region fused to the hinge region, and (iii) an immunoglobulin heavy chain CH3 constant region fused to the CH2 constant region.
- the binding domain polypeptide can be a heavy chain variable region or a light chain variable region.
- the binding-domain immunoglobulin fusion proteins are further disclosed in US 2003/0118592 and US 2003/0133939. These antibody fragments are obtained using conventional techniques known to those skilled in the art, and the fragments are screened for utility in the same manner as are intact antibodies.
- antigen-binding fragments are so-called microantibodies, which are derived from single CDRs.
- microantibodies which are derived from single CDRs.
- Heap et al., 2005 describe a 17 amino acid residue microantibody derived from the heavy chain CDR3 of an antibody directed against the gpl20 envelope glycoprotein of HIV-1 (Heap C.J. et al. (2005); J. Gen. Virol. 86: 1791-1800).
- Other examples include small antibody mimetics comprising two or more CDR regions that are fused to each other, preferably by cognate framework regions.
- antibody or antigen-binding fragment thereof refers to immunoglobulin molecules and immunologically active portions of immunoglobulin molecules, i.e. molecules that contain an antigen-binding site that immunospecifically binds an antigen. Also comprised are immunoglobulin-like proteins that are selected through techniques including, for example, phage display to specifically bind to a target molecule or target epitope.
- the immunoglobulin molecules of the invention can be of any type (e.g., IgG, IgE, IgM, IgD, IgA and IgY), class (e.g., IgGl, IgG2, preferably IgG2a and IgG2b, IgG3, IgG4, IgAl and IgA2) or subclass of immunoglobulin molecule.
- type e.g., IgG, IgE, IgM, IgD, IgA and IgY
- class e.g., IgGl, IgG2, preferably IgG2a and IgG2b, IgG3, IgG4, IgAl and IgA2
- subclass of immunoglobulin molecule e.g., IgG, IgE, IgM, IgD, IgA and IgY
- subclass of immunoglobulin molecule e.g., IgG, IgE, IgM, Ig
- Antibodies and antigen-binding fragments thereof usable in the invention may be from any animal origin including birds and mammals.
- the antibodies or fragments are from human, chimpanzee, rodent (e.g. mouse, rat, guinea pig, or rabbit), chicken, turkey, pig, sheep, goat, camel, cow, horse, donkey, cat, or dog origin. It is particularly preferred that the antibodies are of human or murine origin.
- Antibodies of the invention also include chimeric molecules in which an antibody constant region derived from one species, preferably human, is combined with the antigen-binding site derived from another species, e.g. mouse.
- antibodies of the invention include humanized molecules in which the antigen-binding sites of an antibody derived from a non-human species (e.g. from mouse) are combined with constant and framework regions of human origin.
- antibodies of the invention can be obtained directly from hybridomas which express the antibody, or can be cloned and recombinantly expressed in a host cell (e.g., a CHO cell, or a lymphocytic cell).
- a host cell e.g., a CHO cell, or a lymphocytic cell.
- host cells are microorganisms, such as E. coli, and fungi, such as yeast.
- they can be produced recombinantly in a transgenic non-human animal or plant.
- chimeric antibody refers to those antibodies wherein one portion of each of the amino acid sequences of heavy and light chains is homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular class, while the remaining segment of the chain is homologous to corresponding sequences in another species or class.
- the variable region of both light and heavy chains mimics the variable regions of antibodies derived from one species of mammals, while the constant portions are homologous to sequences of antibodies derived from another.
- One clear advantage to such chimeric forms is that the variable region can conveniently be derived from presently known sources using readily available B-cells or hybridomas from non-human host organisms in combination with constant regions derived from, for example, human cell preparations.
- variable region has the advantage of ease of preparation and the specificity is not affected by the source, the constant region being human is less likely to elicit an immune response from a human subject when the antibodies are injected than would the constant region from a nonhuman source.
- definition is not limited to this particular example.
- humanized antibody refers to a molecule having an antigen-binding site that is substantially derived from an immunoglobulin from a non-human species, wherein the remaining immunoglobulin structure of the molecule is based upon the structure and/or sequence of a human immunoglobulin.
- the antigen-binding site may either comprise complete variable domains fused onto constant domains or only the complementarity determining regions (CDR) grafted onto appropriate framework regions in the variable domains.
- CDR complementarity determining regions
- Antigen-binding sites may be wild-type or modified by one or more amino acid substitutions, e.g. modified to resemble human immunoglobulins more closely.
- Some forms of humanized antibodies preserve all CDR sequences (for example a humanized mouse antibody which contains all six CDRs from the mouse antibody). Other forms have one or more CDRs which are altered with respect to the original antibody.
- human antibodies include antibodies having variable and constant regions derived from human germline immunoglobulin sequences.
- the human antibodies of the invention may include amino acid residues not encoded by human germline immunoglobulin sequences (e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo).
- Human antibodies of the invention include antibodies isolated from human immunoglobulin libraries or from animals transgenic for one or more human immunoglobulin and that do not express endogenous immunoglobulins, as described for example in U.S. Patent No. 5,939,598 by Kucherlapati & Jakobovits.
- the term “monoclonal antibody” as used herein refers to a preparation of antibody molecules of single molecular composition.
- a monoclonal antibody displays a single binding specificity and affinity for a particular epitope.
- the monoclonal antibodies are produced by a hybridoma which includes a B cell obtained from a non-human animal, e.g. mouse, fused to an immortalized cell.
- recombinant antibody includes all antibodies that are prepared, expressed, created or isolated by recombinant means, such as (a) antibodies isolated from an animal (e.g., a mouse) that is transgenic or transchromosomal with respect to the immunoglobulin genes or a hybridoma prepared therefrom, (b) antibodies isolated from a host cell transformed to express the antibody, e.g. from a transfectoma, (c) antibodies isolated from a recombinant, combinatorial antibody library, and (d) antibodies prepared, expressed, created or isolated by any other means that involve splicing of immunoglobulin gene sequences to other DNA sequences.
- transfectoma includes recombinant eukaryotic host cells expressing an antibody, such as CHO cells, NS/0 cells, HEK293 cells, HEK293T cells, plant cells, or fungi, including yeast cells.
- a “heterologous antibody” is defined in relation to a transgenic organism producing such an antibody. This term refers to an antibody having an amino acid sequence or an encoding nucleic acid sequence corresponding to that found in an organism not consisting of the transgenic organism, and being generally derived from a species other than the transgenic organism.
- heterohybrid antibody refers to an antibody having light and heavy chains of different organismal origins.
- an antibody having a human heavy chain associated with a murine light chain is a heterohybrid antibody.
- nanobodies also known as single domain antibodies
- anti-idiotypic antibodies including, e.g., anti-Id antibodies to antibodies described herein
- epitope-binding fragments of any of the above include, e.g., anti-Id antibodies to antibodies described herein, and epitope-binding fragments of any of the above.
- the antibodies described herein are preferably isolated.
- An “isolated antibody” as used herein, is intended to refer to an antibody which is substantially free of other antibodies having different antigenic specificities (e.g., an isolated antibody that specifically binds to C5a is substantially free of antibodies that specifically bind antigens other than C5a).
- An isolated antibody that specifically binds to an epitope, isoform or variant of human C5a may, however, have cross-reactivity to other related antigens, e.g. from other species (e.g. C5a species homologs, such as rat C5a).
- an isolated antibody may be substantially free of other cellular material and/or chemicals.
- a combination of “isolated” monoclonal antibodies relates to antibodies having different specificities and being combined in a well-defined composition.
- naturally occurring refers to the fact that an object can be found in nature.
- a polypeptide or polynucleotide sequence that is present in an organism (including viruses) that can be isolated from a source in nature and which has not been intentionally modified by man in the laboratory is naturally occurring.
- Vilobelimab (sometimes referred to as IFX-1; InflaRx GmbH, Germany) is an antibody specifically binding to C5a.
- the CDR sequences and FR sequences of IFX-1 are disclosed in WO 2011/063980 Al (in Table 4 on page 51), the content of which is hereby incorporated by reference in its entirety.
- the affinity (Ka) and blocking activity is disclosed in WO 2011/063980 Al (in Table 3 on pages 49-50), the content of which is hereby incorporated by reference in its entirety.
- BNJ364, BNJ367, BNJ371, BNJ378, BNJ366, BNJ369, BNJ381 and BNJ383 are antibodies specifically binding to C5a.
- the CDR sequences and FR sequences of these antibodies are disclosed in WO 2011/137395 Al (in Table 2 starting on page 55), the content of which is incorporated by reference in its entirety.
- MEDI-7814 (Medlmmune) is a recombinant humanized anti-C5a antibody.
- the crystal structure of the human C5a in complex with MEDI7814 is available in the RCSB Protein Data Bank under 4UU9 (DOI: 10.2210/pdb4uu9/pdb).
- Sarilumab (brand name Kevzara) is a further antibody binding to IL-6R that is suitable to exercise the invention.
- Sarilumab is a human monoclonal antibody binding to the IL-6 receptor.
- Levilimab is a fully human monoclonal antibody acting against the interleukin-6 receptor (IL-6R), developed by Biocad, for the treatment of rheumatoid arthritis.
- IL-6R interleukin-6 receptor
- Levilimab targets membrane bound and soluble IL-6 receptor.
- Vobarilizumab (synonym ALX-0061) is an high affinity anti-IL-6R bispecific nanobody further targeting albumin to extend half-life.
- Siltuximab brand name Sylvant
- IL-6 interleukin-6
- Siltuximab is FDA-approved for treatment of patients with idiopathic multicentric Castleman’s disease (iMCD) who do not have human immunodeficiency virus (HIV) or human herpesvirus-8 (HHV-8).
- iMCD idiopathic multicentric Castleman’s disease
- HAV human immunodeficiency virus
- HHV-8 human herpesvirus-8
- Olokizumab is a humanized monoclonal antibody specifically binding to IL-6. Olokizumab has been tested for treatment of rheumatoid arthritis and has been used as an emergency experimental cytokine storm COVID-19 treatment.
- Elsilimomab is a monoclonal antibody that targets and blocks IL-6. Elsilimomab has been under development for treatment of lymphoma and myeloma.
- Clazakizumab is a humanized monoclonal antibody directed against IL-6. Clazakizumab has been under development for treatment of psoriatic arthritis.
- PF-4236921 (sometimes referred to as PF-04236921) is an anti-IL-6 monoclonal antibody that has been under development for the treatment of rheumatoid arthritis, systemic lupus erythematosus and Crohn's disease.
- EBL031 (sometimes referred to as RG-6179) is a humanised monoclonal antibody that specifically binds to IL-6.
- a “patient” means any mammal or bird who may benefit from a treatment with the compound described herein (i.e. with an inhibitor of C5a activity described herein).
- a “patient” is selected from the group consisting of laboratory animals (e.g. mouse or rat), domestic animals (including e.g. guinea pig, rabbit, chicken, turkey, pig, sheep, goat, camel, cow, horse, donkey, cat, or dog), or primates including chimpanzees and human beings. It is particularly preferred that the “patient” is a human being.
- treat means accomplishing one or more of the following: (a) reducing the severity and/or duration of the disorder; (b) limiting or preventing development of symptoms characteristic of the disorder(s) being treated; (c) inhibiting worsening of symptoms characteristic of the disorder(s) being treated; (d) limiting or preventing recurrence of the disorder(s) in patients that have previously had the disorder(s); and (e) limiting or preventing recurrence of symptoms in patients that were previously symptomatic for the disorder(s).
- “prevent”, “preventing”, “prevention”, or “prophylaxis” of a disease or disorder means preventing that a disorder occurs in subject.
- an “effective amount” is an amount of a therapeutic agent sufficient to achieve the intended purpose.
- the effective amount of a given therapeutic agent will vary with factors such as the nature of the agent, the route of administration, the size and species of the animal to receive the therapeutic agent, and the purpose of the administration.
- the effective amount in each individual case may be determined empirically by a skilled artisan according to established methods in the art.
- active agent refers to any therapeutic activity an agent may exhibit.
- “Pharmaceutically acceptable” means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans.
- carrier refers to a diluent, adjuvant, excipient, or vehicle with which the therapeutic agent is administered.
- Such pharmaceutical carriers can be sterile liquids, such as saline solutions in water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like.
- a saline solution is a preferred carrier when the pharmaceutical composition is administered intravenously.
- Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions.
- Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like.
- the composition if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents. These compositions can take the form of solutions, suspensions, emulsions, tablets, pills, capsules, powders, sustained-release formulations and the like.
- the composition can be formulated as a suppository, with traditional binders and carriers such as triglycerides.
- the compounds of the invention can be formulated as neutral or salt forms.
- Pharmaceutically acceptable salts include those formed with free amino groups such as those derived from hydrochloric, phosphoric, acetic, oxalic, tartaric acids, etc., and those formed with free carboxyl groups such as those derived from sodium, potassium, ammonium, calcium, ferric hydroxides, isopropylamine, triethylamine, 2-ethylamino ethanol, histidine, procaine, etc.
- suitable pharmaceutical carriers are described in "Remington's Pharmaceutical Sciences" by E. W. Martin.
- Such compositions will contain a therapeutically effective amount of the compound, preferably in purified form, together with a suitable amount of carrier so as to provide the form for proper administration to the patient.
- the formulation should suit the mode of administration.
- the present invention provides an inhibitor of C5a activity for use in:
- the inhibitor of C5a activity and the inhibitor of IL-6 activity are preferably administered within the half-life of the other inhibitor.
- the inhibitor of IL-6 activity would be administered within about 3 days after the administration of the inhibitor of C5a activity or if the inhibitor of IL-6 activity is administered first, the inhibitor of C5a activity is administered within 13 days after the inhibitor of IL-6 activity.
- an overlap between the action of both inhibitors should exist in combination treatment.
- the inhibitor of C5a activity and the inhibitor of IL-6 activity are preferably administered within 1-5 (i.e. 1, 2, 3, 4 or 5) half-lives of the other inhibitor.
- the inhibitor of C5a activity and the inhibitor of IL-6 activity are administered on the same day.
- the inhibitor of IL-6 activity is administered within 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 days prior or after the administration of the inhibitor of C5a activity.
- the inhibitor of C5a activity is administered within 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 days prior or after the administration of the inhibitor of IL-6 activity.
- the patients benefiting most from the use of the inhibitor of C5a activity in the treatment of infectious pneumonia and/or infectious acute respiratory distress syndrome (ARDS) are patients that are or have been treated with an inhibitor of IL-6 activity.
- the inhibitor of IL-6 activity is or has been administered to the patient within a time-window so that there is an overlap between the action of the inhibitor of IL-6 activity and the inhibitor of C5a activity, at least with regard to the administration of the first dose of the inhibitor of C5a activity.
- the present invention provides an inhibitor of IL-6 activity for use in:
- the inhibitor of IL-6 activity and the inhibitor of C5a activity are preferably administered within the half-life of the other inhibitor.
- the inhibitor of IL-6 activity would be administered within about 4 days after the administration of the inhibitor of C5a activity or if the inhibitor of IL-6 activity is administered first, the inhibitor of C5a activity is administered within 13 days after the inhibitor of IL-6 activity.
- an overlap between the action of both inhibitors should exist in combination treatment.
- the inhibitor of IL-6 activity and the inhibitor of C5a activity are administered on the same day.
- the inhibitor of IL-6 activity is administered within 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 days after the administration of the inhibitor of C5a activity.
- the inhibitor of C5a activity is administered within 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 days after the administration of the inhibitor of IL-6 activity.
- the patients benefiting most from the use of the inhibitor of IL-6 activity in the treatment of infectious pneumonia and/or infectious acute respiratory distress syndrome (ARDS) are patients that are or have been treated with an inhibitor of C5a activity.
- the inhibitor of C5a activity is or has been administered to the patient within a time-window so that there is an overlap between the action of the inhibitor of IL-6 activity and the inhibitor of C5a activity, at least with regard to the administration of the first dose of the inhibitor of IL-6 activity.
- the inhibitor of IL-6 activity specifically binds to IL-6 or the IL-6 receptor.
- the inhibitor of IL-6 activity comprises a protein ligand.
- the inhibitor of IL-6 activity is a protein ligand.
- the inhibitor of IL-6 activity is an antibody or antigen binding fragment thereof specifically binding to the IL-6 receptor.
- the inhibitor of IL-6 activity is an antibody or antigen binding fragment thereof specifically binding to IL-6.
- the inhibitor of IL- 6 activity is specifically binding to IL-6R, preferably selected from Tocilizumab, Sarilumab, Levilimab and Vobarilizumab.
- IL-6R preferably selected from Tocilizumab, Sarilumab, Levilimab and Vobarilizumab.
- the inhibitor of IL-6R is Tocilizumab.
- the inhibitor of IL- 6 activity is specifically binding to IL-6R, preferably selected from Siltuximab, Sirukumab, Olokizumab, Elsilimomab, Clazakinumab, PF-423691 and EBI-031.
- the inhibitor of C5a activity binds specifically to C5a or to the C5a receptor (C5aR). In a preferred embodiment, the inhibitor of C5a activity binds specifically to C5a. In a preferred embodiment, the inhibitor of C5a activity binds specifically to C5aR. In a preferred embodiment the inhibitor of C5a activity comprises a protein ligand specifically binding to C5a or the C5aR. In a preferred embodiment the inhibitor of C5a activity comprises a protein ligand specifically binding to C5a. In a preferred embodiment the inhibitor of C5a activity comprises a protein ligand specifically binding to C5aR.
- the inhibitor of C5a activity is a small molecule C5aR antagonist.
- Suitable small molecule C5aR antagonists are known in the art and can inter alia be found in WO 2020/182384 Al, which is incorporated by reference herein. Methods for preparing the small molecule C5aR antagonists can be found in WO 2020/182384 AL
- the inhibitor of C5a activity is a small molecule having the general formula (I) and pharmaceutically acceptable salts, hydrates and rotamers thereof; wherein
- C 1 is selected from the group consisting of aryl and heteroaryl, wherein the heteroaryl group has from 1-3 heteroatoms as ring members selected from N, O and S; and wherein said aryl and heteroaryl groups are optionally substituted with from 1 to 3 R 1 substituents;
- C 2 is selected from the group consisting of aryl and heteroaryl, wherein the heteroaryl group has from 1-3 heteroatoms as ring members selected from N, O and S; and wherein said aryl and heteroaryl groups are optionally substituted with from 1 to 3 R 2 substituents;
- C 3 is selected from the group consisting of Ci-8 alkyl or heteroalkyl, C3-8 cycloalkyl, C3-8 cycloalkyl-Ci-4 alkyl, aryl, aryl-Ci-4 alkyl, heteroaryl, heteroaryl-Ci-4 alkyl, heterocycloalkyl or heterocycloalkyl-Ci-4 alkyl, wherein the heteroalkyl group has from 1-3 heteroatoms selected from N, O and S, wherein the heterocycloalkyl group or portion has from 1-3 heteroatoms selected from N, O and S, and wherein the heteroaryl group has from 1-3 heteroatoms as ring members selected from N, O and S, and each C 3 is optionally substituted with from 1 to 3 R 3 substituents; each R 1 is independently selected from the group consisting of halogen, — CN, -R c , — CO 2 R a , — CONR a R b , — C(O)R a
- X is hydrogen or CH3; and R 8 and R 9 are independently from each other selected from the group consisting of hydrogen, halogen, Ci-Cs alkyl, Ci-Cs haloalkyl, and Ci-Cs alkoxy or R 8 and R 9 are combined to form a fused saturated or unsaturated mono- or multi-ring carbocycle in which one or more of the ring carbon atoms may be replaced independently from each other by N, S, or O, with the proviso that at least one of R 8 and R 9 is not hydrogen.
- the inhibitor of C5a activity is a small molecule C5aR antagonist having the general formula (I) as indicated above and wherein R 8 and/or R 9 is hydrogen.
- the inhibitor of C5a activity is selected from:
- INF 004 (2R,3 S)-2-(4-(cyclopentylamino)phenyl)- 1 -(2-fluoro-6-methylbenzoyl)-
- INF022 (2R,3S)-2-(4-(cyclopentylamino)phenyl)-6-fluoro-l-(2-fluoro-6- methylbenzoyl)-N-(4-methyl-3-(tri fluoromethyl)phenyl)- 1,2,3, 4-tetrahydroquinoline-3- carb oxami de
- INF023 (2R,3S)-2-(4-(cyclopentylamino)phenyl)-l-(2-fluoro-6-methylbenzoyl)-
- INF024 (2R,3S)-2-(4-(cyclopentylamino)phenyl)-l-(2-fluoro-6-methylbenzoyl)-
- INF030 (2R, 3 S)- 1 -(2-fluoro-6-methylbenzoyl)-N-(4-(hydroxymethyl)-3 -
- INF034 (2R, 3 S)- 1 -(2-fluoro-6-methylbenzoyl)-2-(4-(3 -hydroxy-3 - methylbutyl)phenyl)-N-(4-methyl-3-(trifluoromethyl)phenyl)-l,2,3,4-tetrahydroquinoline-3- carb oxami de
- INF035 (2R,3S)-2-(4-(cyclopentylamino)phenyl)-l-(2-fluoro-6-methylbenzoyl)-
- INF041 (2R,3S)-2-(4-(cyclopentylamino)phenyl)-l-(2-fluorobenzoyl)-N-(4-
- INF048 (2R,3S)-l-(2-fluoro-6-methylbenzoyl)-2-(4-(3-hydroxy-3- methylbutyl)phenyl)-N-(4-(hydroxymethyl)-3-(trifluoromethyl)phenyl)-l,2,3,4- tetrahydroquinoline-3-carboxamide
- INF050 (2R,3S)-2-(4-(cyclobutylamino)phenyl)-l-(2-fluoro-6-methylbenzoyl)-
- INF052 (2R,3S,5R)-2-(4-(cyclopentylamino)phenyl)-l-(2,6-difluorobenzoyl)-
- INF053 (2R,3S,5R)-2-(4-(cyclopentylamino)phenyl)-l-(2,6-dimethylbenzoyl)-
- INF055 (2R,3S)-2-(4-(cyclopentylamino)phenyl)-l-(2-fluoro-6-methylbenzoyl)-
- INF056 (2R,3S,5R)-2-(4-(cyclopentylamino)phenyl)-l-(2-fluoro-6- methylbenzoyl)-N-(4-methyl-3-(trifluoromethyl)phenyl)-5-(trifluoromethyl)piperidine-3- carb oxami de
- INF068 (2R,3S,5R)-2-(4-(cyclopentylamino)phenyl)-l-(2,6-difluorobenzoyl)-
- INF069 (2R,3S,5R)-2-(4-(cyclopentylamino)phenyl)-l-(2,6-difluorobenzoyl)-
- INF070 (2R,3S,5R)-l-(2,6-difluorobenzoyl)-N-(4-methyl-3-
- INF072 (2R,3S,5R)-N-(3-chloro-4-hydroxyphenyl)-2-(4-
- INF077 (2R,3S,5R)-N-(4-chloro-3-(trifluoromethyl)phenyl)-2-(4-
- INF080 (2R,3S,5R)-2-(4-(cyclopentylamino)phenyl)-N-(3,4-dichlorophenyl)-l-
- the inhibitor of C5a activity is INF052.
- the inhibitor of C5a activity is avacopan, having the structure of general formula (II):
- the inhibitor of C5a activity is a protein ligand, wherein the protein ligand is an antibody or an antigen binding fragment thereof. More preferably the protein ligand comprises a VH and a VL domain of an antibody, a Fab fragment, a Fab’ fragment, a heavy chain antibody, a single-domain antibody (sdAb), variable domain of a heavy chain antibody, a VHH, a nanobody, a single-chain variable fragment (scFv), a tandem scFv, or a single-chain diabody.
- sdAb single-domain antibody
- scFv single-chain variable fragment
- scFv tandem scFv
- the inhibitor of C5a activity is a protein ligand, wherein the protein ligand is an antibody-like protein, preferably an affibody, anticalin, or an designed ankyrin repeat protein.
- Antibody-like proteins include without limitation affibodies, anticalins, and designed ankyrin repeat proteins (for review see: Binz H.K. et al. (2005) Engineering novel binding proteins from non-immunoglobulin domains. Nat. Biotechnol. 23(10): 1257-1268).
- Antibody-like proteins can be derived from large libraries of mutants, e.g. be panned from large phage display libraries and can be isolated in analogy to regular antibodies. Also, antibody-like binding proteins can be obtained by combinatorial mutagenesis of surface-exposed residues in globular proteins. Antibody-like proteins are sometimes referred to as “peptide aptamers”.
- the inhibitor of C5a activity is a protein ligand, wherein the protein ligand is an inhibitory variant of C5a.
- the inhibitor of C5a activity is a protein ligand, wherein the protein ligand is an inhibitory variant of C5a receptor.
- the inhibitor of C5a activity is a protein ligand, preferably an antibody or antigen-binding fragment thereof, specifically binding to a conformational epitope of human C5a formed by (a) amino acid sequences NDETCEQRA (SEQ ID NO: 2) and SHKDMQL (SEQ ID NO: 3) of C5a binds to a conformational epitope of C5a formed by amino acid sequences NDETCEQRA (SEQ ID NO: 2) and SHKDMQL (SEQ ID NO: 3), and binds to at least one amino acid within the amino acid sequence according to SEQ ID NO: 2 and to at least one amino acid within the amino acid sequence according to SEQ ID NO: 3.
- NDETCEQRA SEQ ID NO: 2
- SHKDMQL SEQ ID NO: 3
- the inhibitor of C5a activity according to this preferred embodiment binds at the same time to at least one amino acid within the amino acid sequence according to SEQ ID NO: 2 and to at least one amino acid within the amino acid sequence according to SEQ ID NO: 3.
- SEQ ID NO: 2 corresponds to amino acids 30-38 of human C5a.
- SEQ ID NO: 3 corresponds to amino acids 66-72 of human C5a.
- the amino acid sequence of human C5a is depicted in SEQ ID NO: 1.
- the inhibitor of C5a activity binds to at least one of amino acids DETCEQR (SEQ ID NO: 4).
- SEQ ID NO: 4 corresponds to amino acids 31-37 of human C5a.
- the inhibitor of C5a activity binds to at least one of amino acids HKDMQ (SEQ ID NO: 5), more preferably to at least one of amino acids KDM.
- SEQ ID NO: 5 correspond to amino acids 67-71 of human C5a; the sequence KDM corresponds to amino acids 68-70 of human C5a.
- the inhibitor of C5a activity binds at the same time to at least one amino acid within the amino acid sequence DETCEQR (SEQ ID NO: 4) and to at least one amino acid within the amino acid sequence KDM.
- the specific binding to the amino acids of the conformational epitope is determined by alanine scanning. In a preferred embodiment the conformational epitope is determined by alanine scanning.
- the spatial epitopes containing peptide C5a 28-40 (VNNDETCEQRAAR, SEQ ID NO: 53) and C5a peptide 65-70 (ISHKDM, SEQ ID NO: 54) can be viewed as random coils.
- the two peptides are linked by a flexible peptide linker, GGGGS (SEQ ID NO: 55)
- the spatial epitopes is reconstructed resembling the parent antigen conformation, as the weak hydrophobic interaction from the two peptides ensures a pocket-shape conformation.
- the inhibitor of C5a activity is an antibody or antigen-binding fragment thereof specifically binding to C5a comprising a variable domain of a heavy chain (VH) and a variable domain of a light chain (VL).
- VH variable domain of a heavy chain
- VL variable domain of a light chain
- the VL comprises a light chain CDR3 sequence CQQSNEDPYT as set forth in SEQ ID NO: 6.
- the VL further comprises a FR2 sequence WYQQKPGQPPKLL as set forth in SEQ ID NO: 8 or SEQ ID NO: 39.
- the VL comprises a light chain CDR3 sequence CQQNNEDPLT as set forth in SEQ ID NO: 7.
- the VL further comprises a FR2 sequence WYQQKPGQPPKLL as set forth in SEQ ID NO: 8 or SEQ ID NO: 39.
- the inhibitor of C5a activity further comprises at least one of the following sequences:
- the inhibitor of C5a activity comprises a light chain CDR3 sequence according to SEQ ID NO: 6, a light chain CDR2 sequence according to SEQ ID NO:
- the inhibitor of C5a activity further comprises the light chain framework regions (FR) 1 to 4 according to SEQ ID NOs: 37, 39, 41 and 43, respectively; preferably further comprising the heavy chain FR 1 to 4 according to SEQ ID NOs:45, 47, 49 and 51, respectively.
- FR light chain framework regions
- the inhibitor of C5a activity comprises a light chain CDR3 sequence according to SEQ ID NO: 7, a light chain CDR2 sequence according to SEQ ID NO:
- the inhibitor of C5a activity further comprises the light chain framework regions (FR) 1 to 4 according to SEQ ID NOs:28, 8, 29 and 30, respectively; preferably further comprising the heavy chain FR 1 to 4 according to SEQ ID NOs:32, 33, 34 and 35, respectively.
- FR light chain framework regions
- the FR sequences have a sequence identity to the herein disclosed FR sequences of at least 99%, at least 98% at least 95%, at least 90%, at least 85%, at least 80% or at least 75% sequence.
- the inhibitor of C5a activity is selected from IFX-1 (vilobelimab), IFX-2, INab708, BNJ364, BNJ367, BNJ371, BNJ378, BNJ366, BNJ369, BNJ381, BNJ383, MEDI-7814 or ALXN-1007, or an antigenbinding fragment thereof.
- the inhibitor of C5a activity is IFX-1 (vilobelimab).
- the inhibitor of C5a activity is selected from an antibody or antigen binding fragment thereof that competes for binding with any of the antibodies selected from:IFX-l (vilobelimab), IFX-2, INab708, BNJ364, BNJ367, BNJ371, BNJ378, BNJ366, BNJ369, BNJ381, BNJ383, MEDI-7814 or ALXN-1007.
- the inhibitor of C5a activity is an antibody or antigen-binding fragment thereof that specifically binds to human C5a comprises (i) a light chain variable domain (VL) having at least 99%, at least 98% at least 95%, at least 90%, at least 85%, at least 80%, at least 75% sequence identity with the VL domain according to SEQ ID NO: 22 and (ii) a heavy chain variable domain (VH) having at least 99%, at least 98% at least 95%, at least 90%, at least 85%, at least 80%, at least 75% sequence identity with the VH domain according to SEQ ID NO: 27.
- the sequence variation does not include the CDR sequences.
- the inhibitor of C5a activity is an antibody or antigen-binding fragment thereof that specifically binds to human C5a comprises (i) a light chain variable domain (VL) according to SEQ ID NO: 22 and (ii) a heavy chain variable domain (VH) according to SEQ ID NO: 27.
- the inhibitor of C5a activity is an antibody or antigen-binding fragment thereof that specifically binds to human C5a comprises (i) a light chain variable domain (VL) having at least 99%, at least 98% at least 95%, at least 90%, at least 85%, at least 80%, at least 75% sequence identity with the VL domain according to SEQ ID NO: 31 and (ii) a heavy chain variable domain (VH) having at least 99%, at least 98% at least 95%, at least 90%, at least 85%, at least 80%, at least 75% sequence identity with the VH domain according to SEQ ID NO: 36.
- the sequence variation does not include the CDR sequences.
- the inhibitor of C5a activity is an antibody or antigen-binding fragment thereof that specifically binds to human C5a comprises (i) a light chain variable domain (VL) having at least 99%, at least 98% at least 95%, at least 90%, at least 85%, at least 80%, at least 75% sequence identity with the VL domain according to SEQ ID NO: 44 and (ii) a heavy chain variable domain (VH) having at least 99%, at least 98% at least 95%, at least 90%, at least 85%, at least 80%, at least 75% sequence identity with the VH domain according to SEQ ID NO: 52.
- the sequence variation does not include the CDR sequences.
- the inhibitors of C5a activity are antibodies or antigen binding fragments thereof disclosed herein comprising one, two or three modified amino acids within the CDR sequences.
- the modified amino acids are selected from substitution, insertion or deletion, preferably substitution.
- the one, two or three modified amino acids are conservative amino acid substitutions. “Conservative substitutions” may be made, for instance, on the basis of similarity in polarity, charge, size, solubility, hydrophobicity, hydrophilicity, and/or the amphipathic nature of the amino acid residues involved.
- Amino acids can be grouped into the following six standard amino acid groups:
- hydrophobic Met, Ala, Vai, Leu, He
- neutral hydrophilic Cys, Ser, Thr, Asn, Gin
- conservative amino acid substitutions are defined as exchanges of an amino acid by another amino acid listed within the same group of the six standard amino acid groups shown above. For example, the exchange of Asp by Glu retains one negative charge in the so modified polypeptide.
- glycine and proline may be substituted for one another based on their ability to disrupt a-helices.
- Some preferred conservative substitutions within the above six groups are exchanges within the following sub-groups: (i) Ala, Vai, Leu and He; (ii) Ser and Thr; (ii) Asn and Gin; (iv) Lys and Arg; and (v) Tyr and Phe. Given the known genetic code, and recombinant and synthetic DNA techniques, the skilled scientist readily can construct DNAs encoding the conservative amino acid variants.
- inhibitors of C5a activity disclosed herein may freely be combined with the inhibitors of IL-6 activity for the claimed use.
- the inhibitor of C5a activity is vilobelimab and the inhibitor of IL-6 activity is tocilizumab.
- Pneumonia is a medical condition of the lung characterized by inflammation of the lung tissue in particular the aleveoli. Pneumonia can be caused by various pathogens, including bacteria, viruses, fungi, and other microorganisms. The infection causes the alveoli in the lungs to become filled with fluid, pus, and cellular debris, which impairs the lungs' ability to efficiently exchange oxygen and carbon dioxide.
- infectious pneumonia is viral pneumonia, bacterial pneumonia or fungal pneumonia, preferably viral pneumonia or bacterial pneumonia, more preferably viral pneumonia.
- Viral pneumonia may be caused by various viruses including but not limited to influenza virus A or B, respiratory syncytial virus, and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2).
- SARS-CoV-2 severe acute respiratory syndrome coronavirus 2
- infectious viral pneumonia is caused by SARS-CoV-2.
- the infectious pneumonia is COVID-19 pneumonia, i.e. viral pneumonia caused by SARS-CoV-2 infection.
- Acute respiratory distress syndrome (ARDS) is a severe and life-threatening condition that affects the lungs. ARDS is characterized by inflammation and damage to the alveoli. This damage results in increased permeability of the alveolar-capillary membrane, causing fluid leakage into the alveoli impairing their ability to effectively exchange oxygen and carbon dioxide.
- the ARDS is viral ARDS or bacterial ARDS. In a more preferred embodiment the viral ARDS is COVID-19 ARDS.
- the inhibitor of C5a activity or the inhibitor of IL-6 activity are for administration in a regime comprising an induction phase followed by a maintenance phase.
- the inhibitor of C5a activity is administered in the induction phase and the maintenance phase and the inhibitor of IL-6 activity is administered:
- an additional second dose of the inhibitor of IL-6 activity is administered according to (I), (II) or (III).
- the inhibitor of C5a activity is administered in the induction phase and the maintenance phase and the inhibitor of IL-6 activity is administered within 1 to 4 weeks, preferably within 1-2 weeks, more preferably within 1 week, prior to the induction phase.
- the inhibitor of C5a activity is administered in the induction phase and the maintenance phase and the inhibitor of IL-6 activity is administered 1 to 14 days, preferable 1 to 7 days, prior to the induction phase.
- the inhibitor of C5a activity is administered in the induction phase and the maintenance phase and the inhibitor of IL-6 activity is administered during the induction phase.
- the inhibitor of C5a activity is administered in the induction phase and the maintenance phase and the inhibitor of IL-6 activity is administered during the maintenance phase.
- the inhibitor of C5a activity is administered before the inhibitor of IL-6 activity
- the induction phase has a duration of 1 to 10 days, preferably 2 to 8 days, more preferably 8 days.
- 1 to 8 doses preferably 2 to 6 doses, more preferably 3 to 5 doses, most preferably 4 doses, of the inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab) are administered.
- the inhibitor of C5a activity preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab
- each dose administered in the induction phase comprises 200-1600mg, preferably 400-1200mg, more preferably 600-1000mg, most preferably 800mg, of the inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab).
- the inhibitor of C5a activity preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab.
- the inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab) is administered by intravenous infusion or injection, preferably infusion.
- the maintenance phase has a duration of 1 to 4 weeks, preferably 2-3 weeks, most preferably 2 weeks.
- the inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab) is administered every 1 to 4 weeks, preferably 1 to 2 weeks, more preferably every week.
- each dose administered in the maintenance phase comprises 200-1600mg, preferably 400-1200mg, more preferably 600-1000mg, most preferably 800mg, of the inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab).
- the inhibitor of C5a activity preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab.
- the inhibitor of C5a activity and the inhibitor of IL-6 activity are used in an administration regimen, wherein
- the induction phase comprises or essentially consists of a period of 1 to 10 days, preferably 2 to 8 days, more preferably 7-8 days, wherein 1-8 doses, preferably 2-4 doses, comprising each 200-1600 mg, preferably 400-1200 mg of said inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab), are administered; and
- the maintenance phase comprises or essentially consists of a period of at least 1 week, preferably 2-4 weeks, wherein a dose, comprising 200-1600 mg, preferably 400-1200 mg of said inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab), is administered every 1-4 weeks, preferably every week.
- a dose comprising 200-1600 mg, preferably 400-1200 mg of said inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab)
- the inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab) is administered within the induction and the maintenance phase at a total maximum of 4 to 12 doses, preferably 6 to 10 doses, more preferably 6 to 8 doses, most preferably 6 doses.
- the inhibitor of C5a activity and the inhibitor of IL-6 activity are used in an administration regimen, wherein
- the induction phase essentially consists of a period of 8 days, wherein 4 doses comprising each 800 mg of the inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab), are administered (preferably on days 1, 2, 4 and 8); and
- the maintenance phase essentially consists of a period of 2 weeks, wherein a dose, comprising 800 mg of the inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab), is administered every 1 week (preferably on day 15 and 22 of the treatment).
- the inhibitor of IL-6 activity (preferably tocilizumab) is administered at a dose of 200-1600mg per dose, preferably 400-1200mg, more preferably 600- lOOOmg, most preferably 800mg.
- the inhibitor of IL-6 activity (preferably tocilizumab) is administered at a dose of 2 mg/kg to 12 mg/kg, preferably 3 mg/kg to 10 mg/kg, more preferably 4 mg/kg to 8 mg/kg.
- the inhibitor of IL-6 activity (preferably tocilizumab) is administered at a weight dependent dose of 8mg/kg for patients at or above 30 kg weight or 12 mg/kg for patients of less than 30 kg weight.
- the inhibitor of IL-6 activity (preferably tocilizumab) is administered at a maximum dose of 800 mg per administration.
- the inhibitor of IL-6 activity (preferably tocilizumab) is administered in 1 to 4 doses, preferably 1-2 doses.
- the inhibitor of IL-6 activity (preferably tocilizumab) is administered as a single dose.
- the inhibitor of IL-6 activity (preferably tocilizumab) is administered as a first and second dose.
- a second dose is in particular beneficial if clinical signs or symptoms do not improve after the first dose.
- the additional dose is at administered at least 8 hours after the first dose.
- each further dose of the inhibitor of IL-6 activity is administered at least 8 hours after the previous administration.
- an additional dose of the inhibitor of IL-6 activity is administered 8-24 hours after the first dose.
- the inhibitor of IL-6 activity is administered every 1 to 4 weeks, preferably every 1 to 2 weeks, preferably weekly.
- the inhibitor of IL-6 activity (preferably tocilizumab) is administered by intravenous infusion or injection, preferably infusion.
- each dose of the inhibitor of IL-6 activity (preferably tocilizumab) is administered prior to the induction phase.
- the inhibitor of IL-6 activity (preferably tocilizumab) is administered as 1-2 doses, preferably single dose, prior (preferably 1-2 weeks prior, more preferably within 1 week prior) to the induction phase.
- the inhibitor of IL-6 activity (preferably tocilizumab) is administered within the induction phase.
- the inhibitor of IL-6 activity (preferably tocilizumab) is administered in more than a single dose, wherein at least one dose is administered prior to the induction phase and at least one dose is administered in the induction phase and/or maintenance phase.
- the inhibitor of IL-6 activity (preferably tocilizumab) is administered in more than a single dose, wherein at least one dose is administered prior to the induction phase and at least one dose is administered in the induction phase.
- the inhibitor of IL-6 activity (preferably tocilizumab) is administered in more than a single dose, wherein at least one dose is administered in the induction phase and at least one dose is administered in the maintenance phase.
- the inhibitor of C5a activity and the inhibitor of IL-6 activity are used in an administration regimen, wherein
- 1 or 2 doses (preferably single dose) of the inhibitor of IL-6 activity is administered within 0 to 14 days prior and/or after the start of the induction phase (preferably within 0 to 7 days (i.e. prior or after) of the start of the induction phase, more preferably within 24 hours (i.e. prior or after) of the start of the induction phase), preferably the inhibitor of IL-6 activity (preferably tocilizumab) is administered at a weight dependent dose of 8mg/kg for patients at or above 30 kg weight or 12 mg/kg for patients of less than 30 kg weight;
- the induction phase comprises or essentially consists of a period of 1 to 10 days, preferably 2 to 8 days more preferably 7-8 days, wherein 1-8 doses, preferably 2-4 doses, comprising each 200-1600 mg, preferably 400-1200 mg of the inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab), are administered; and
- the maintenance phase comprises or essentially consists of a period of at least 2 weeks, preferably 2-4 weeks, wherein a dose, comprising 200-1600 mg, preferably 400-1200 mg of said inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab), is administered every 1-4 weeks, preferably every week.
- a dose comprising 200-1600 mg, preferably 400-1200 mg of said inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab)
- a dose comprising 200-1600 mg, preferably 400-1200 mg of said inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab) is administered every 1-4 weeks, preferably every week.
- the inhibitor of C5a activity and the inhibitor of IL-6 activity are used in an administration regimen, wherein
- the induction phase comprises or essentially consists of a period of 1 to 10 days, preferably 2 to 5 days, wherein 1-8 doses, preferably 2-4 doses, comprising each 200-1600 mg, preferably 400-1200 mg of the inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab), are administered;
- the maintenance phase comprises or essentially consists of a period of at least 2 weeks, preferably 2-4 weeks, wherein a dose, comprising 200-1600 mg, preferably 400-1200 mg of said inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab), is administered every 1-4 weeks, preferably every week.
- a dose comprising 200-1600 mg, preferably 400-1200 mg of said inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab)
- a dose comprising 200-1600 mg, preferably 400-1200 mg of said inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab) is administered every 1-4 weeks, preferably every week.
- the inhibitor of C5a activity and the inhibitor of IL-6 activity are used in an administration regimen, wherein
- the induction phase comprises or essentially consists of a period of 1 to 10 days, preferably 2 to 5 days, wherein 1-8 doses, preferably 2-4 doses, comprising each 200-1600 mg, preferably 400-1200 mg of the inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab), are administered;
- the maintenance phase comprises or essentially consists of a period of at least 2 weeks, preferably 2-4 weeks, wherein a dose, comprising 200-1600 mg, preferably 400-1200 mg of said inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab), is administered every 1-4 weeks, preferably every week.
- a dose comprising 200-1600 mg, preferably 400-1200 mg of said inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab)
- the inhibitor of C5a activity and the inhibitor of IL-6 activity are used in an administration regimen, wherein
- the induction phase comprises or essentially consists of a period of 1 to 10 days, preferably 2 to 5 days, wherein 1-8 doses, preferably 2-4 doses, comprising each 200-1600 mg, preferably 400-1200 mg of the inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab), are administered;
- the maintenance phase comprises or essentially consists of a period of at least 2 weeks, preferably 2-4 weeks, wherein a dose, comprising 200-1600 mg, preferably 400-1200 mg of said inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab), is administered every 1-4 weeks, preferably every week; and
- the inhibitor of C5a activity and the inhibitor of IL-6 activity are used in an administration regimen, wherein
- 1 or 2 doses (preferably single dose) of the inhibitor of IL-6 activity is administered within 0 to 14 days prior and/or after the start of the induction phase (preferably within 0 to 7 days (i.e. prior or after) of the start of the induction phase);
- the induction phase comprises or essentially consists of a period of 1 to 10 days, preferably 2 to 5 days, wherein 1-8 doses, preferably 2-4 doses, comprising each 200-1600 mg, preferably 400-1200 mg of the inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab), are administered;
- the maintenance phase comprises or essentially consists of a period of at least 2 weeks, preferably 2-4 weeks, wherein a dose, comprising 200-1600 mg, preferably 400-1200 mg of said inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab), is administered every 1-4 weeks, preferably every week; and
- the inhibitor of C5a activity and the inhibitor of IL-6 activity are used in an administration regimen, wherein (i) the induction phase comprises or essentially consists of a period of 1 to 10 days, preferably 2 to 5 days, wherein 1-8 doses, preferably 2-4 doses, comprising each 200-1600 mg, preferably 400-1200 mg of the inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab), are administered;
- the maintenance phase comprises or essentially consists of a period of at least 2 weeks, preferably 2-4 weeks, wherein a dose, comprising 200-1600 mg, preferably 400-1200 mg of said inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab), is administered every 1-4 weeks, preferably every week; and
- the inhibitor of C5a activity and the inhibitor of IL-6 activity are used in an administration regimen, wherein
- 1 or 2 doses (preferably single dose) of the inhibitor of IL-6 activity is administered within 0 to 14 days prior and/or after the start of the induction phase (preferably within 0 to 7 days (i.e. prior or after) of the start of the induction phase);
- the induction phase comprises or essentially consists of a period of 1 to 10 days, preferably 2 to 5 days, wherein 1-8 doses, preferably 2-4 doses, comprising each 200-1600 mg, preferably 400-1200 mg of the inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab), are administered;
- the maintenance phase comprises or essentially consists of a period of at least 2 weeks, preferably 2-4 weeks, wherein a dose, comprising 200-1600 mg, preferably 400-1200 mg of said inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab), is administered every 1-4 weeks, preferably every week; and
- the inhibitor of C5a activity and the inhibitor of IL-6 activity are used in an administration regimen, wherein
- 1 dose of the inhibitor of IL-6 activity is administered within 24 hours (i.e. prior or after) of the first administration of the inhibitor of C5a activity (preferably vilobelimab); optionally a second dose of the inhibitor of IL-6 activity (preferably tocilizumab) is administered, preferably after at least 8 hours of the first dose of the inhibitor of IL-6 activity; wherein the inhibitor of IL-6 activity (preferably tocilizumab) is administered at a weight dependent dose of 8mg/kg for patients at or above 30 kg weight or 12 mg/kg for patients of less than 30 kg weight; (ii) the inhibitor of C5a activity (preferably vilobelimab) is administered on day 1, day 2, day 4, day 8, day 15 and day 22, wherein the inhibitor of C5a activity (preferably vilobelimab) is administered at a dose of 800 mg.
- the inhibitor of C5a activity or the inhibitor of IL-6 activity are for use in treating patients that are on invasive mechanical ventilation, preferably not more than 48h post intubation upon first administration of the inhibitor of C5a activity.
- the inhibitor of C5a activity or the inhibitor of IL-6 activity are for use in treating patients that have a Path / FiCL ratio of between 50 and 300, preferably between 60 and 200.
- PaCL/FiCL ratio is used inter alia to classify severity of acute respiratory distress syndrome (ARDS). In a preferred embodiment the following classification of ARDS severity is used: mild (PaCL/FiCL ratio 200-300), moderate (PaCL/FiCL ratio 100-200) and severe (PaCL/FiCL ratio ⁇ 100).
- the inhibitor of C5a activity or the inhibitor of IL-6 activity are for use in treating patients with mild to severe (preferably moderate to severe) ARDS.
- the present invention provides a kit of parts comprising an inhibitor of C5a activity (preferably vilobelimab) and an inhibitor of IL-6 activity (preferably tocilizumab).
- the kit is further comprising instructions for the use of the inhibitor of C5a activity (preferably vilobelimab) and the inhibitor of IL-6 activity (preferably tocilizumab) and/or means for the application of the compounds.
- the kit of parts comprises the inhibitor of C5a activity (preferably vilobelimab) in a dosage form comprising 100-2000 mg, 200-1500mg, 400- 1200mg, preferably 600-1000 mg, more preferably 800mg of an inhibitor of C5a activity (preferably vilobelimab).
- the dosage form is a lyophilized powder, or a liquid dosage form, preferably a solution for infusion or a solution for injection.
- the kit of parts comprises the inhibitor of IL-6 activity (preferably tocilizumab) in a dosage form comprising 100-2000 mg, 200-15 OOmg, 400- 1200mg, preferably 600-1000 mg, more preferably 800mg of an inhibitor of IL-6 activity (preferably tocilizumab).
- the dosage form is a lyophilized powder, or a liquid dosage form, preferably a solution for infusion or a solution for injection.
- a method for the treatment of patients with an infectious pneumonia and/or infectious acute respiratory distress syndrome comprising administering to the patient an effective amount of an inhibitor of C5a activity and an inhibitor of IL-6 activity.
- the method comprises administering the inhibitor of IL-6 activity prior and/or concomitant with the inhibitor of C5a- activity.
- the present invention provides the use of an inhibitor of C5a activity and an inhibitor of IL-6 activity for the manufacturing of a medicament for the treatment of infectious pneumonia and/or infectious acute respiratory distress syndrome (ARDS).
- ARDS infectious acute respiratory distress syndrome
- Example 1 Phase II/III study evaluating IFX-1 for the treatment of COVID 19 related severe pneumonia
- Neutrophil driven tissue and organ damage is known to play an important role in a wide array of acute inflammatory diseases.
- the mechanism leading to damage has been largely attributed to two mechanisms: 1) the release of granular enzymes and 2) the generation of so-called reactive oxygen species in which O2 radical formation elicits a damaging effect.
- Phase II of the current trial was exploratory in nature and was not powered to show statistically significant differences in clinical endpoints.
- Relative change (%) from baseline to day 5 in the oxygenation index, chosen as the primary endpoint for Phase II showed a large variability and dependency on patient positioning and intubation status which excluded this endpoint from being used in a confirmatory study.
- Phase III of the study is an adequately powered, placebo-controlled, double blinded phase evaluating standard of care (SOC) + IFX-1 versus SOC + placebo-to-match with 28-day all-cause mortality as the primary endpoint, an accepted regulatory primary endpoint for critical care studies.
- SOC standard of care
- the SOC to be utilized in Phase III of this study reflects the current understanding of the SOC for hospitalized patients with COVID-19, which includes venous thromboembolism (VTE) prophylaxis at a minimum, and may include other international and country-specific recommended treatments for COVID-19 per the locally adopted treatment recommendations.
- VTE venous thromboembolism
- the aim for determining the dose and administration frequency for IFX-1 is to establish a pragmatic administration schedule for the COVID-19 patient population.
- IFX-1 dose and administration schedule was chosen based on prior pharmacokinetic (PK)/pharmacodynamic (PD) observations for IFX-1 trough values and blood C5a levels, dose-response assessment in Study IFX-1 in Hidradentis suppurativa, and the described unprecedented high C5a levels in patients with severe COVID-19 related pneumonia (Gao et al. 2020; Carvelli et al. 2020).
- the dosing schedule of IFX-1 for this study includes an additional dose of 800 mg IFX 1 at day 2 in the established fractionated loading dose scheme, that foresees administration of 800 mg at days 1, 4, and 8.
- This additional dose has been chosen due to the reported high C5a levels in COVID-19 patients (Gao et al. 2020). Therefore, the entire dosing scheme contains up to 6 IFX-1 doses of 800 mg at days 1, 2, 4, 8, 15, and 22 (or less until hospital discharge). This regimen had been employed in Phase II of this study, leading to a lower death rate and supporting efficacy signals without any new signals of unknown toxicities (Vlaar et al 2020).
- Phase III The primary objective of Phase III is:
- Phase III is a double-blind, placebo-controlled, randomized phase evaluating SOC + IFX-1 (Arm A) and SOC + placebo- to-match (Arm B).
- the SOC includes VTE prophylaxis at a minimum, and may include other international and country-specific recommended treatments for COVID-19 per the locally adopted treatment recommendations.
- Phase III of the study will randomize up to 400 patients with one interim analysis for stopping for futility.
- a total of 180 patients are planned to be randomized into Arm A and Arm B using a 1 : 1 allocation ratio for the first stage; based on results of the interim analysis, up to an additional 180 patients are planned to be randomized using the same allocation ratio for the second stage. Additional patients will be randomized under the following conditions:
- the primary statistical analysis will be based on all randomized patients except patients randomized in error who did not get IMP treatment.
- Phase III patients will be treated with a maximum of 6 IV doses of IFX-1 800 mg (Arm A) or placebo (Arm B) at days 1, 2, 4, 8, 15, and 22, as long as the patient is still in the hospital (even if discharged from the ICU).
- End of treatment is defined as the last planned administration or discontinuation of IMP for other reasons.
- the FUV is scheduled to occur 28 days (if patient has been discharged) and 60 days after the randomization date.
- the visit information obtained at a site visit or via telephone call e.g., AEs, survival status, according to the SoA should be recorded.
- the end of study for the individual patient is defined as the date of the last contact of the follow up period (i.e., 60 days after randomization), date of death, date of consent withdrawal from study participation, or the date of last contact when patients are lost to followup, whatever occurs earliest.
- treatment of COVID 19 with investigational treatments which are not approved or not included in locally adopted treatment guidelines for this indication e.g., WHO guidance, NIH COVID-19 treatment guidelines
- SARS CoV-2 multiplication inhibitor(s) or immunomodulator(s) e.g., SARS CoV-2 multiplication inhibitor(s) or immunomodulator(s).
- a locally adopted treatment guideline recommends drugs such as remdesivir, dexamethasone, or anticoagulation, this would be allowed.
- Adopted guidelines and updates must be documented at study initiation and throughout the conduct of the study.
- Screen failures Screening failures are defined as patients who do not meet the criteria for participation in this study and thus, are either not randomized/assigned to IMP or randomized in error and do/did not receive IMP. Screen failure data for not randomized patients will not be recorded in the eCRF, for those patients who are randomized in error, screen failure data are to be recorded in the eCRF.
- the IMP is defined as the investigational treatment IFX-1 and Placebo-to-match (Placebo) that is intended to be administered to a study patient according to the study protocol.
- IFX-1 concentrate solution for infusion will be supplied in 20 mL glass vials at a concentration of 10 mg/mL (200 mg per vial) for reconstitution and IV administration.
- the solution will contain sodium chloride, sodium phosphate, and polysorbate 80.
- Placebo concentrate solution for infusion will be supplied in 20 mL glass vials for reconstitution and IV administration.
- the solution will contain sodium chloride, sodium phosphate, and polysorbate 80.
- Phase III patients are randomized to either receive SOC + IFX-1 in treatment Arm A or SOC + Placebo in treatment Arm B.
- the first dose of IFX-1 or Placebo is administered at Day 1.
- SOC may start at any time according to their administration schedule.
- Patients will be treated with a maximum of 6 IV doses of IFX-1 800 mg (Arm A) or placebo (Arm B) administered as a 30-60-minute IV infusion.
- the 6 treatments are at days 1, 2, 4, 8, 15, and 22 as long as the patient is still in the hospital, even if discharged from ICU.
- IMP administrations for Phase III are detailed in Figure 1.
- SOC treatment start and stop is not defined by the protocol and can start at any time. SOC treatment will be given according to investigator’s discretion.
- Reportable medications or vaccinations that the patient is receiving within 7 days prior to randomization and up to 30 days after the last dose of study treatment should be recorded along with:
- prior and concomitant medications will follow special considerations for conditions in the ICU.
- a complete record of all prior and concomitant medications (but excluding nutritional and volume therapy, electrolyte support, vitamins, non-steroidal antiinflammatory drugs (NSAIDs), and supportive therapies such as artificial tears, ointments, stool softeners/laxatives, etc.) will be maintained in the eCRF for each participant, beginning 7 days before randomization and continuing up to 30 days after the last dose of study treatment).
- NSAIDs non-steroidal antiinflammatory drugs
- An AE is any untoward medical occurrence in a subject administered an IMP; an AE does not necessarily have to have a causal relationship with this treatment.
- AEs encompass any unfavorable and unintended sign (including an abnormal laboratory finding), symptom, or disease that arises or worsens after the inclusion of the subject into the study.
- a total of 180 patients (90 per arm) are planned to be randomized in Stage 1 and up to 180 patients (90 per arm) in Stage 2.
- patients randomized in error (screening failures) and patients for whom consent is withdrawn within 48 hours after randomization additional patients will be randomized.
- the power calculation is based on an overall 2.5% one-sided alpha and an assumed 30% 28-day mortality under Placebo and 15% 28-day mortality under IFX-1 treatment.
- Full analysis set The primary statistical analysis will be based on all randomized patients except patients randomized in error (reason for early termination documented as “Randomized by mistake” in the eCRF) who did not get IMP treatment.
- Safety analysis set SAF: Safety analyses will be based on all patients who received at least one infusion of IMP, and patients will be analyzed according to the treatment they actually received. Statistical Analyses
- the primary analysis will be performed based on the FAS.
- the primary efficacy variable is 28-day mortality (proportion of patients deceased until day 28).
- the primary statistical hypotheses to be tested are:
- HR is the hazard ratio derived from the beta coefficient of the primary statistical analysis Cox regression model comparing the SOC + Placebo treatment arm (Arm B) with the SOC + IFX-1 treatment arm (Arm A).
- a total of 180 patients are planned to be randomized to Arm A and Arm B using a 1 : 1 allocation ratio for the first stage.
- the interim analysis is performed after 180 patients, not counting patients being randomized in error, have been followed-up until day 28 (or died before).
- 180 additional patients are planned to be randomized in a ratio of 1 : 1 to Arm A and B.
- the planned maximum number of patients in the study is 360. Depending on the number of patients meeting criteria for additional randomization, more patients will be randomized but not more than 400.
- the primary statistical analysis of IFX-1 + SOC compared to SOC alone will make use of a one-sided alpha level of 2.5% and will test for superiority (lower mortality among IFX-1 treated patients).
- the primary statistical analysis will be based on a Cox proportional hazards regression model with outcome 28-day all-cause mortality as a censored time-to-event variable and explanatory variables treatment arm (Arm B versus Arm A) and age.
- the primary statistical analysis will be based on all randomized patients except patients randomized in error (reason for early termination documented as “Randomized by mistake” in the eCRF) who did not get IMP treatment.
- all-cause mortality will be censored at day 28 for subjects who died after day 28 or who have more than 28 days follow-up alive.
- the z-statistic for the interim and final analysis will be calculated as the proportion of the beta coefficient for the treatment arm effect and its standard error from the Cox proportional hazards regression model. The study will be stopped for futility if the z-statistic for the first stage is 0 or lower.
- the secondary efficacy analyses will be performed based on the FAS.
- the first secondary efficacy endpoint will only be addressed with statistical hypothesis tests if the primary endpoint is statistically significant using the full overall 2.5% one-sided alpha. If the first secondary endpoint is also statistically significant, the full 2.5% one-sided alpha will be passed to the 4 remaining secondary endpoints. Multiplicity in the secondary endpoints will be addressed with the fallback method. 60-day mortality will be analyzed similar to 28-day mortality and the other secondary endpoints will be evaluated using a logistic regression model with the explanatory variables randomized treatment arm (Arm B versus Arm A) and age for:
- the ordering of the secondary endpoints 2 to 5 for hypothesis testing will be as in above mentioned list.
- the full 2.5% one-sided alpha for the fallback method will be attributed to the 4 secondary endpoints in the following way 2%, 0.2%, 0.2%, and 0.1%. If the preceding hypothesis test is not significant, subsequent tests will be performed at the aforementioned alpha level. If tests are significant, the alpha is added to the subsequent hypothesis test (e.g., if the primary hypothesis test is significant and secondary endpoints 1-4 are all significant, the fifth secondary endpoint will be tested at an alpha of 2.5%; if the fourth secondary endpoint is not significant, the fifth secondary endpoint will be tested at an alpha of 0.1%).
- the primary endpoint as well as all secondary endpoints will also be evaluated as censored time to event variables by Kaplan-Meier type methods. Kaplan-Meier analyses will be performed comparing the two treatment arms overall and within the following stratifications:
- Time to first extubation will be analyzed as a censored time to event variable. Withdrawing early from the study will lead to the patient being right-censored at the date of withdrawal provided that the reason is not an outcome of interest (extubation or death). In order to adequately account for competing outcomes as well as potential right-censoring due to varying follow-up times, the outcome will be non-parametrically analyzed using Aalen- Johansen-type estimation based on a competing risks model with two absorbing states (event 1 : all-cause death; event 2: extubation). Adjustment for age will be realized by regression modeling accounting for competing risks.
- Patients achieving 8-point ordinal scale score 3 or below (patients alive and free of respiratory failure) at day 15 and day 28 are defined as alive and free of respiratory failure at the respective study day.
- the corresponding endpoint will be analyzed using logistic regression with explanatory variables age and treatment group.
- Glasgow Outcome Scale assessed at study day 60 will be analyzed via ordinal logistic regression with explanatory variables treatment arm (Arm B versus Arm A) and age.
- EQ-5D Quality of life will be assessed by EQ-5D at study day 60.
- the visual analogue scale as well as an index value based on the 5 health states (cross-walk index value using the United States value set) will be analyzed by an ANCOVA model with explanatory variables treatment arm (Arm B versus Arm A), age, and sex (male versus female).
- the interim analysis is performed after the first 180 patients, not counting patients being randomized in error, in Stage 1 have been followed-up until day 28 (or died before).
- the study will be stopped for futility if the z-statistic of the primary efficacy analysis is 0 or lower. If the study is not stopped for futility, another up to 180 patients will be randomized in Stage 2. Depending on the number of patients meeting criteria for additional randomization, more than 360 patients will be randomized but not more than a total of 400.
- Example 2 Patients receiving combined treatment of IFX-1 and Tocilizumab in Phase III study evaluating IFX-1 for the treatment of COVID 19 related severe pneumonia
- IFX-1 vilobelimab
- tocilizumab patients receiving both medications, i.e. IFX-1 (vilobelimab) and tocilizumab, exhibited a highly significant reduction of all-cause mortality as compared to placebo treatment as well as single use of vilobelimab or tocilizumab.
- Baseline use of tocilizumab is defined as receiving at least one dose of tocilizumab between study day -7 and study day 1.
- Prior use of tocilizumab is defined as receiving at least one dose of tocilizumab before day-7 (i.e. prior to baseline use) concomitant use: Medications will be counted as concomitant either
- the previous medication will also be assumed to be concomitant. Incomplete start/end dates will not be imputed. If the incomplete date does not allow clear categorization as previous or concomitant medication, the medication will be counted as concomitant.
- tocilizumab use is either classified as ‘baseline usage of Tocilizumab’ or ‘any usage of Tocilizumab’, whereas the later encompasses prior use, baseline use and concomitant use.
- the primary statistical analysis is based on a Cox proportional hazards regression model with outcome 28-day and 60-day all-cause mortality as a censored time-to-event variable and explanatory variables treatment arm (Arm B (i.e. placebo + SOC) versus Arm A (i.e. vilobelimab + SOC) and age. This statistical analysis was made with patients receiving Tocilizumab (baseline use or any use) in Arm A and or Arm B.
- a hazard ratio (HR) above 1 indicates a covariate that is positively associated with the event probability, and thus negatively associated with the length of survival.
- HR 1 : No effect
- HR ⁇ 1 Reduction in the hazard
- HR > 1 Increase in Hazard. Therefore a covariate with a hazard ratio > 1 is a bad prognostic factor and a covariate with a hazard ratio ⁇ 1 is called good prognostic factor.
- the combined treatment of vilobelimab and tocilizumab (any usage) is significantly more effective in reducing all cause mortality as compared to tocilizumab alone.
- Table 2 Cox proportional hazard model for 28-day all-cause mortality for patients with any usage of Tocilizumab - FAS; (number of patients in analysis set/model: 61/61)
- Table 3 Cox proportional hazard model for 60-day all-cause mortality for patients with any usage of Tocilizumab - FAS; (number of patients in analysis set/model: 61/61)
- Tocilizumab defined as receiving at least one dose of tocilizumab between study day -7 and study day 1).
- Table 4 Cox proportional hazard model for 28-day all-cause mortality for patients with baseline usage of Tocilizumab - FAS; (number of patients in analysis set/model: 60/60);
- Table 8 Cox proportional hazard model for 28-day all-cause mortality for patients without baseline usage of Tocilizumab - FAS; (number of patients in analysis set/model: 308/308)
- Table 10 Cox proportional hazards regression model with outcome 28-day all-cause mortality as a censored time-to-event variable and explanatory variables treatment arm and age and baseline tocilizumab use flag and its interaction with study treatment - FAS. (number of patients in analysis set/model: 368/368)
- Table 11 Cox proportional hazards regression model with outcome 60-day all-cause mortality as a censored time-to-event variable and explanatory variables treatment arm and age and baseline tocilizumab use flag and its interaction with study treatment - FAS. (number of patients in analysis set/model: 368/368)
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Organic Chemistry (AREA)
- Medicinal Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Animal Behavior & Ethology (AREA)
- Immunology (AREA)
- Molecular Biology (AREA)
- Pharmacology & Pharmacy (AREA)
- Veterinary Medicine (AREA)
- Virology (AREA)
- Biophysics (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Genetics & Genomics (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Biochemistry (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Oncology (AREA)
- Communicable Diseases (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Pulmonology (AREA)
- Engineering & Computer Science (AREA)
- Epidemiology (AREA)
- Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
- Peptides Or Proteins (AREA)
Abstract
The present invention relates to an inhibitor of C5a activity and an inhibitor of IL-6 activity for use in the treatment of infectious pneumonia and infectious acute respiratory distress syndrome (ARDS). In further aspects administration regimes and kit of parts are referred to.
Description
Treatment of pneumonia and ARDS with inhibitors of C5a and IL-6 activity
The present invention relates to an inhibitor of C5a activity and an inhibitor of IL-6 activity for use in the treatment of infectious pneumonia and infectious acute respiratory distress syndrome (ARDS). In further aspects administration regimes and kit of parts are referred to.
Background of the Invention
C5a
C5a is a 74 amino acid spanning split product of its “mother molecule” C5 and represents one endpoint of the complement activation cascade. It can be generated through activation of at least three well-described pathways (the alternative, the classical and the MBL pathway). All pathways merge at the level of C3, forming the C5- or alternative C5 convertase leading to cleavage of C5 into C5a and C5b. The latter binds with C6, C7, C8 and multiple C9 molecules ultimately leading to formation of pores in e.g. bacterial membranes (terminal Membrane Attack Complex = MAC). C5a is generated when the complement system is activated in settings of inflammation and other immunological and inflammatory disorders / diseases.
Among the complement activation products, C5a is one of the most potent inflammatory peptides, with a broad spectrum of functions (Guo and Ward, 2005). C5a exerts its effects through the high-affinity C5a receptors (C5aR and C5L2) (Ward, 2009). C5aR belongs to the rhodopsin family of G-protein-coupled receptors with seven transmembrane segments; C5L2 has a similar structure but appears not to be G-protein-coupled. It is currently believed that C5a exerts its biological functions primarily through C5a-C5aR interaction. However, some reports demonstrate signaling also through C5L2 activation (Rittirsch and others, 2008).
C5aR is widely expressed on myeloid cells including neutrophils, eosinophils, basophils, and monocytes, and non-myeloid cells in many organs, especially in the lung and liver, indicative of the importance of C5a/C5aR signaling. Widespread up-regulation of C5aR expression occurs during the onset of sepsis, and blockade of C5a/C5aR interaction by anti- C5a, or anti-C5aR antibodies, or C5aR antagonists renders highly protective effects in rodent models of sepsis (Czermak and others, 1999; Huber-Lang and others, 2001; Riedemann and others, 2002).
C5a has been reported to exert numerous pro-inflammatory responses. For example, C5a stimulates the synthesis and release from human leukocytes of pro-inflammatory cytokines
such as TNF-a, IL-ip, IL-6, IL-8, and macrophage migration inhibitory factor (MIF) (Hopken U et al. 1996. Eur J Immunol 26(5): 1103-1109; Riedemann NC et al. 2004. J Immunol 173(2): 1355-1359; Stricter RM et al. 1992. Am J Pathol 141(2):397-407). C5a produces a strong synergistic effect with LPS in production of TNF-a, macrophage inflammatory protein (MIP)-2, cytokine-induced neutrophil chemoattractant (CINC)-l, and IL-ip in alveolar epithelial cells (Riedemann NC et al. 2002. J. Immunol. 168(4): 1919-1925; Rittirsch D et al. 2008. Nat Rev Immunol 8(10):776-787).
C5a has a variety of biological functions (Guo and Ward, 2005). C5a is a strong chemoattractant for neutrophils and also has chemotactic activity for monocytes and macrophages. C5a causes an oxidative burst (02 consumption) in neutrophils and enhances phagocytosis and release of granular enzymes. C5a has also been found to be a vasodilator. C5a has been shown to be involved in modulation of cytokine expression from various cell types and to enhance expression of adhesion molecule expression on neutrophils. High doses of C5a can lead to nonspecific chemotactic “desensitization” of neutrophils, thereby causing broad dysfunction.
Interleukin-6 (IL-6)
IL-6 has a fundamental role in inflammation, immune regulation, hematopoiesis, host defense, homeostasis, and tissue regeneration. It is produced by a wide range of hematopoietic and somatic cells that influences numerous cell types with multiple biological functions.
IL-6 plays a critical role in the immune response by stimulating the growth and differentiation of T and B cells, as well as promoting the production of antibodies. Further, IL- 6 is involved in the acute phase response to injury or infection, promoting inflammation and activating immune cells. Its function in hematopoiesis is to stimulate the production of blood cells in the bone marrow, including red blood cells, white blood cells, and platelets. IL-6 is further involved in the regulation of glucose metabolism and lipid metabolism and has been implicated in various neurological functions, including neuroprotection and neuroinflammation. Furthermore, abnormal IL-6 production has been associated with the development of a wide variety of systemic immune-mediated, chronic diseases, and even neoplasms.
IL-6 transmits its signals through a cell-surface type-I receptor complex, that consists of a ligand-binding glycoprotein termed as IL-6 receptor (IL-6R) and a signal-transducing component gpl30. When IL-6 binds to IL-6R, it induces a conformational change that enables the recruitment and assembly of gp!30. This leads to the formation of a high-affinity IL-6
receptor complex, which activates intracellular signaling pathways. The downstream signaling pathways activated by the IL-6 receptor complex depend on the cell type and context but can include the JAK-STAT pathway, the PI3K-Akt pathway, and the MAPK pathway.
Two forms of IL-6R are known: a membrane-bound form (mIL-6R) and a soluble form (sIL-6R). The membrane-bound form is found on the surface of cells and is responsible for the high-affinity binding of IL-6. The soluble form is generated by proteolytic cleavage of mIL-6R and can bind to IL-6, forming a complex with gpl30 and inducing intracellular signaling. The sIL-6R is capable of activating cells that do not express the membrane-bound form of IL-6R, expanding the range of cells that can respond to IL-6.
The IL-6 receptor is expressed on a wide range of cells, including immune cells, hepatocytes, adipocytes, and neurons.
Corona virus infection
Different corona virus outbreaks occurred in the last decades worldwide of which in particular three types of virus resulted in diseases with high mortality rates. The Severe Acute Respiratory Syndrome (SARS), the Middle East Respiratory Syndrome (MERS) and lately Coronavirus disease 2019 (COVID-19). The coronavirus responsible for these outbreaks where SARS-CoV, MERS-CoV and SARS-CoV-2, respectively.
COVID-19 typically presents with features of long non-symptomatic latency which might be a main contributing factor to a relatively high transmissibility compared to the other previously occurring deadly coronavirus infections, severe acute respiratory syndrome (SARS) and Middle East respiratory syndrome (MERS). COVID-19 patients typically present with flulike symptoms such as fever or signs of lower respiratory tract illness including dry cough and shortness of. With the progression of disease into a severe form, it often affects multiple organs’ functions including the lung, heart, liver, and coagulation system among others. As such, death is typically caused by respiratory failure and multiple organ dysfunctions similar to other viral pneumonia-induced sepsis. Sepsis and ARDS mostly occur in the second week upon disease onset.
Older age, underlying health conditions (e.g. cardiovascular issues) and compromised immune systems are important risk factors for the potential of developing a more severe form of disease onset and worse outcomes. The main comorbidities alongside COVID-19 that are associated with a high death rate are hypertension, diabetes and coronary heart disease.
COVID-19 can be characterized by a dual play of viral inflammation and immune- mediated injury that may result in a “complement storm” event occurring in the progression of corona virus infection, in particular COVID-19.
Anti-C5a therapy for COVID-19 has been disclosed in W02021/190770 Al and recently received emergency use authorization (EUA) by the FDA. Likewise, the use of an IL- 6 receptor inhibitor (Tocilizumab; also known as Actemra) for the treatment of CO VID-19 also received an EUA and BLA from the FDA.
In the present invention the surprising effect of a combined anti-C5a and anti-IL-6 therapy of infectious pneumonia and/or infectious acute respiratory distress syndrome (ARDS) is disclosed.
Summary of the Invention
In a first aspect, the present invention provides an inhibitor of C5a activity for use in:
(i) combination with an inhibitor of IL-6 activity in the treatment of infectious pneumonia and/or infectious acute respiratory distress syndrome (ARDS); and/or
(ii) the treatment of infectious pneumonia and/or infectious acute respiratory distress syndrome (ARDS), wherein the patient is or has been treated with an inhibitor of IL-6 activity.
In a second aspect, the present invention provides an inhibitor of IL-6 activity for use in:
(i) combination with an inhibitor of C5a activity in the treatment of infectious pneumonia and/or infectious acute respiratory distress syndrome (ARDS); and/or
(ii) the treatment of infectious pneumonia and/or infectious acute respiratory distress syndrome (ARDS), wherein the patient is or has been treated with an inhibitor of C5a activity.
In a third aspect, the present invention provides a kit of parts comprising an inhibitor of C5a activity and an inhibitor of IL-6 activity.
List of Figures
In the following, the content of the figures comprised in this specification is described. In this context please also refer to the detailed description of the invention above and/or below.
Figure 1: IMP Administrations in Phase III refers to the administration schedule of the investigational medicinal product (IMP). The anti-C5a antibody IFX-1 (vilobelimab) was administered at a dose of 800mg on several days during the treatment period as indicated in figure 1.
Figure 2: 60-day all-cause mortality for treatment with vilobelimab and any use of tocilizumab. Line 1 : Vilo + SOC with any use of tocilizumab; Line 2: Vilo + SOC without any use of tocilizumab; Line 3: Placebo + SOC with any use of tocvilizumab; Line 4: Placebo + SOC without any use of tocilizumab;
(SOC = Standard of care, Vilo = Vilobelimab)
Figure 3: 60-day all-cause mortality for treatment with vilobelimab and baseline use of tocilizumab. Line 1 : Vilo + SOC with baseline use of tocilizumab; Line 2: Vilo + SOC without baseline use of tocilizumab; Line 3: Placebo + SOC with any use of tocilizumab; Line 4: Placebo + SOC without baseline use of tocilizumab;
(SOC = Standard of care, Vilo = Vilobelimab)
Detailed Descriptions of the Invention
Before the present invention is described in detail below, it is to be understood that this invention is not limited to the particular methodology, protocols and reagents described herein as these 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 invention which will be limited only by the appended claims. Unless defined otherwise, all technical and scientific terms used herein have the same meanings as commonly understood by one of ordinary skill in the art.
Preferably, the terms used herein are defined as described in "A multilingual glossary of biotechnological terms: (IUPAC Recommendations)", Leuenberger, H.G.W, Nagel, B. and Klbl, H. eds. (1995), Helvetica Chimica Acta, CH-4010 Basel, Switzerland).
Throughout this specification and the claims which follow, unless the context requires otherwise, the word "comprise", and variations such as "comprises" and "comprising", will be understood to imply the inclusion of a stated integer or step or group of integers or steps but not the exclusion of any other integer or step or group of integers or steps. In the following passages, different aspects of the invention are defined in more detail. Each aspect so defined may be combined with any other aspect or aspects unless clearly indicated to the contrary. In particular, any feature indicated as being optional, preferred or advantageous may be combined with any other feature or features indicated as being optional, preferred or advantageous.
Several documents are cited throughout the text of this specification. Each of the documents cited herein (including all patents, patent applications, scientific publications, manufacturer's specifications, instructions etc.), whether supra or infra, is hereby incorporated by reference in its entirety. Nothing herein is to be construed as an admission that the invention
is not entitled to antedate such disclosure by virtue of prior invention. Some of the documents cited herein are characterized as being “incorporated by reference ” . In the event of a conflict between the definitions or teachings of such incorporated references and definitions or teachings recited in the present specification, the text of the present specification takes precedence.
In the following, the elements of the present invention will be described. These elements are listed with specific embodiments; however, it should be understood that they may be combined in any manner and in any number to create additional embodiments. The variously described examples and preferred embodiments should not be construed to limit the present invention to only the explicitly described embodiments. This description should be understood to support and encompass embodiments which combine the explicitly described embodiments with any number of the disclosed and/or preferred elements. Furthermore, any permutations and combinations of all described elements in this application should be considered disclosed by the description of the present application unless the context indicates otherwise.
Definitions
In the following, some definitions of terms frequently used in this specification are provided. These terms will, in each instance of its use, in the remainder of the specification have the respectively defined meaning and preferred meanings.
As used in this specification and the appended claims, the singular forms "a", "an", and "the" include plural referents, unless the content clearly dictates otherwise.
The term “about” when used in connection with a numerical value is meant to encompass numerical values within a range having a lower limit that is 5% smaller than the indicated numerical value and having an upper limit that is 5% larger than the indicated numerical value.
Sequences: All sequences referred to herein are disclosed in the attached sequence listing that, with its whole content and disclosure, is a part of this specification.
In the context of the present invention, C5a particularly refers to human C5a. Human C5a is a 74 amino acid peptide with the following amino acid sequence:
TLQKKIEEIA AKYKHSWKK CCYDGACVNN DETCEQRAAR ISLGPRCIKA FTECCWASQ LRANISHKDM QLGR (SEQ ID NO: 1).
The amino acid sequence of human C5 can be found under the accession number UniProtKB P01031 (C05 HUMAN).
As used herein, the term “inhibitor of C5a activity” refers to any compound that in any way reduces the activity of C5a. This activity reduction can be achieved by directly or indirectly lowering the concentration of C5a, or by reducing the activity of C5a, or by preventing that C5a exerts its effects on one or more of its receptors (e.g. on C5aR or C5L2), or by reducing the concentration or activity of one or more receptors of C5a. Preferred inhibitors of C5a activity are antibodies specifically binding to hC5a.
In the context of the present invention, the expression “C5a receptor” refers to any potential C5a binding ligand on the cell surface, especially to any receptor protein to which C5a may bind and elicit a reaction on said receptor (e.g. activation or inhibition of the receptor). The term “C5a receptor” particularly encompasses the two receptors C5aR and C5L2. Alternative names for C5aR are C5aRl and CD88. An alternative name for C5L2 is C5aR2.
In the context of the present invention, Interleukin-6 (IL-6) particularly refers to human IL-6. Human IL-6 is an 212 amino acid peptide. The amino acid sequence of human IL-6 can be found under the accession number UniProtKB P05231 (IL6 HUMAN).
As used herein, the term “inhibitor of IL-6 activity” refers to any compound that in any way reduces the activity of IL-6. This activity reduction can be achieved by directly or indirectly lowering the concentration of IL-6, or by reducing the activity of IL-6, or by preventing that IL-6 exerts its effects on one or more of its receptors (e.g. on IL-6R), or by reducing the concentration or activity of one or more receptors of IL-6. Preferred inhibitors of IL-6 activity are antibodies specifically binding to IL-6 or IL-6R, preferably IL-6R.
In the context of the present invention, the expression “IL-6 receptor” refers to any potential IL-6 binding ligand, especially to any receptor protein to which IL-6 may bind and elicit a reaction on said receptor (e.g. activation or inhibition of the receptor). The term “IL-6 receptor” (IL-6R; sometimes referred to as CD 126) particularly encompasses the two receptor forms sIL-6R and mIL-6R.
As used herein, a first compound (e.g. an antibody or antigen-binding fragment thereof) is considered to “bind” to a second compound (e.g. a target protein), if it has a dissociation constant Ka to said second compound of 1 mM or less, preferably 100 pM or less, preferably 50 pM or less, preferably 30 pM or less, preferably 20 pM or less, preferably 10 pM or less, preferably 5 pM or less, more preferably 1 pM or less, more preferably 900 nM or less, more preferably 800 nM or less, more preferably 700 nM or less, more preferably 600 nM or less, more preferably 500 nM or less, more preferably 400 nM or less, more preferably 300 nM or less, more preferably 200 nM or less, even more preferably 100 nM or less, even more preferably 90 nM or less, even more preferably 80 nM or less, even more preferably 70 nM or
less, even more preferably 60 nM or less, even more preferably 50 nM or less, even more preferably 40 nM or less, even more preferably 30 nM or less, even more preferably 20 nM or less, and even more preferably 10 nM or less.
The term “binding” according to the invention preferably relates to a specific binding. “Specific binding” means that a compound (e.g an antibody or antigen-binding fragment thereof) binds stronger to a target such as an epitope for which it is specific compared to the binding to another target. A compound binds stronger to a first target compared to a second target, if it binds to the first target with a dissociation constant (Ka) which is lower than the dissociation constant for the second target. Preferably the dissociation constant (Ka) for the target to which the compound binds specifically is more than 10-fold, preferably more than 20- fold, more preferably more than 50-fold, even more preferably more than 100-fold, 200-fold, 500-fold or 1000-fold lower than the dissociation constant (Ka) for the target to which the compound does not bind specifically.
As used herein, the term “Ka” (usually measured in “mol/L”, sometimes abbreviated as “M”) is intended to refer to the dissociation equilibrium constant of the particular interaction between a binding moiety (e.g. an antibody or antigen-binding fragment thereof) and a target molecule (e.g. an antigen or epitope thereof).
Methods for determining binding affinities of compounds, i.e. for determining the dissociation constant Ka, are known to a person of ordinary skill in the art and can be selected for instance from the following methods known in the art: Surface Plasmon Resonance (SPR) based technology, Bio-layer interferometry (BLI), enzyme-linked immunosorbent assay (ELISA), flow cytometry, isothermal titration calorimetry (ITC), analytical ultracentrifugation, radioimmunoassay (RIA or IRMA) and enhanced chemiluminescence (ECL). Typically, the dissociation constant Ka is determined at 20°C, 25°C, 30°C, or 37°C. If not specifically indicated otherwise, the Ka values recited herein are determined by surface plasmon resonance spectroscopy (Biacore™) at room temperature (25°C).
Typically, antibodies and antigen-binding fragments thereof according to the invention bind with a sufficient binding affinity to their target, for example, with a Kd value of between 500 nM-1 pM, i.e. 500 nM, 450 nM, 400nM, 350 nM, 300nM, 250 nM, 200nM, 150 nM, lOOnM, 50 nM, 10 nM, 1 nM, 900 pM, 800 pM, 700 pM, 600 pM, 500 pM, 400 pM, 300 pM, 200 pM, 100 pM, 50 pM, IpM.
An “epitope”, also known as antigenic determinant, is the part of a macromolecule that is recognized by the immune system, specifically by antibodies, B cells, or T cells. As used herein, an “epitope” is the part of a macromolecule capable of binding to a compound (e.g. an
antibody or antigen-binding fragment thereof) as described herein. In this context, the term “binding” preferably relates to a specific binding. Epitopes usually consist of chemically active surface groupings of molecules such as amino acids or sugar side chains and usually have specific three-dimensional structural characteristics, as well as specific charge characteristics. Conformational and non-conformational epitopes can be distinguished in that the binding to the former but not the latter is lost in the presence of denaturing solvents.
A “paratope” is the part of an antibody that binds to the epitope.
The term “antibody” typically refers to a glycoprotein comprising at least two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds, or an antigen-binding portion thereof. The term “antibody” also includes all recombinant forms of antibodies, in particular of the antibodies described herein, e.g. antibodies expressed in prokaryotes, unglycosylated antibodies, antibodies expressed in eukaryotes (e.g. CHO cells), glycosylated antibodies, and any antigen-binding antibody fragments and derivatives as described below. Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as VH or VH) and a heavy chain constant region. Each light chain is comprised of a light chain variable region (abbreviated herein as VL or VL) and a light chain constant region. The VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR). Each VH and VL is composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4. The variable regions of the heavy and light chains contain a binding domain that interacts with an antigen. The constant regions of the antibodies may mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the first component (Clq) of the classical complement system.
The term "CDR" or "complementarity determining region" refers to the non-contiguous antigen binding sites found within the variable region of both heavy and light chain polypeptides (sometimes referred to as LCDR or HCDR). CDRs have been described by Kabat et al., J. Biol. Chem. 252:6609-6616 (1977); Kabat et al., U.S. Dept, of Health and Human Services, "Sequences of proteins of immunological interest" (1991) (also referred to herein as Kabat 1991); by Chothia et al., J. Mol. Biol. 196:901-917 (1987) (also referred to herein as Chothia 1987); and MacCallum et al., J. Mol. Biol. 262:732-745 (1996), where the definitions include overlapping or subsets of amino acid residues when compared against each other. Nevertheless, application of either definition to refer to a CDR of an antibody or grafted
antibodies or variants thereof is intended to be within the scope of the term as defined and used herein. The CDRs listed in Table 1 were defined in accordance with Kabat 1991.
The term “antigen-binding fragment” of an antibody (or simply “binding portion”), as used herein, refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen. It has been shown that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody. Examples of binding fragments encompassed within the term “antigen-binding portion” of an antibody include (i) Fab fragments, monovalent fragments consisting of the VL, VH, CL and CH domains; (ii) F(ab')2 fragments, bivalent fragments comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) Fd fragments consisting of the VH and CH domains; (iv) Fv fragments consisting of the VL and VH domains of a single arm of an antibody, (v) dAb fragments (Ward et al., (1989) Nature 341 : 544-546), which consist of a VH domain; (vi) isolated complementarity determining regions (CDR), and (vii) combinations of two or more isolated CDRs which may optionally be joined by a synthetic linker. Furthermore, although the two domains of the Fv fragment, VL and VH, are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv); see e.g., Bird et al. (1988) Science 242: 423-426; and Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85: 5879- 5883). Such single chain antibodies are also intended to be encompassed within the term “antigen-binding fragment” of an antibody. A further example is a binding-domain immunoglobulin fusion protein comprising (i) a binding domain polypeptide that is fused to an immunoglobulin hinge region polypeptide, (ii) an immunoglobulin heavy chain CH2 constant region fused to the hinge region, and (iii) an immunoglobulin heavy chain CH3 constant region fused to the CH2 constant region. The binding domain polypeptide can be a heavy chain variable region or a light chain variable region. The binding-domain immunoglobulin fusion proteins are further disclosed in US 2003/0118592 and US 2003/0133939. These antibody fragments are obtained using conventional techniques known to those skilled in the art, and the fragments are screened for utility in the same manner as are intact antibodies. Further examples of “antigen-binding fragments” are so-called microantibodies, which are derived from single CDRs. For example, Heap et al., 2005, describe a 17 amino acid residue microantibody derived from the heavy chain CDR3 of an antibody directed against the gpl20 envelope glycoprotein of HIV-1 (Heap C.J. et al. (2005); J. Gen. Virol. 86: 1791-1800). Other examples include small antibody mimetics comprising two or more CDR regions that are fused to each other, preferably by cognate framework regions. Such a small antibody mimetic comprising VH CDR1 and VL
CDR3 linked by the cognate VH FR2 has been described by Qiu et al., 2007 (Qiu X.Q. et al. (2007); Nature biotechnology 25(8):921-929).
Thus, the term “antibody or antigen-binding fragment thereof’, as used herein, refers to immunoglobulin molecules and immunologically active portions of immunoglobulin molecules, i.e. molecules that contain an antigen-binding site that immunospecifically binds an antigen. Also comprised are immunoglobulin-like proteins that are selected through techniques including, for example, phage display to specifically bind to a target molecule or target epitope. The immunoglobulin molecules of the invention can be of any type (e.g., IgG, IgE, IgM, IgD, IgA and IgY), class (e.g., IgGl, IgG2, preferably IgG2a and IgG2b, IgG3, IgG4, IgAl and IgA2) or subclass of immunoglobulin molecule.
Antibodies and antigen-binding fragments thereof usable in the invention may be from any animal origin including birds and mammals. Preferably, the antibodies or fragments are from human, chimpanzee, rodent (e.g. mouse, rat, guinea pig, or rabbit), chicken, turkey, pig, sheep, goat, camel, cow, horse, donkey, cat, or dog origin. It is particularly preferred that the antibodies are of human or murine origin. Antibodies of the invention also include chimeric molecules in which an antibody constant region derived from one species, preferably human, is combined with the antigen-binding site derived from another species, e.g. mouse. Moreover, antibodies of the invention include humanized molecules in which the antigen-binding sites of an antibody derived from a non-human species (e.g. from mouse) are combined with constant and framework regions of human origin.
As exemplified herein, antibodies of the invention can be obtained directly from hybridomas which express the antibody, or can be cloned and recombinantly expressed in a host cell (e.g., a CHO cell, or a lymphocytic cell). Further examples of host cells are microorganisms, such as E. coli, and fungi, such as yeast. Alternatively, they can be produced recombinantly in a transgenic non-human animal or plant.
The term “chimeric antibody” refers to those antibodies wherein one portion of each of the amino acid sequences of heavy and light chains is homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular class, while the remaining segment of the chain is homologous to corresponding sequences in another species or class. Typically the variable region of both light and heavy chains mimics the variable regions of antibodies derived from one species of mammals, while the constant portions are homologous to sequences of antibodies derived from another. One clear advantage to such chimeric forms is that the variable region can conveniently be derived from presently known sources using readily available B-cells or hybridomas from non-human host organisms in
combination with constant regions derived from, for example, human cell preparations. While the variable region has the advantage of ease of preparation and the specificity is not affected by the source, the constant region being human is less likely to elicit an immune response from a human subject when the antibodies are injected than would the constant region from a nonhuman source. However, the definition is not limited to this particular example.
The term “humanized antibody” refers to a molecule having an antigen-binding site that is substantially derived from an immunoglobulin from a non-human species, wherein the remaining immunoglobulin structure of the molecule is based upon the structure and/or sequence of a human immunoglobulin. The antigen-binding site may either comprise complete variable domains fused onto constant domains or only the complementarity determining regions (CDR) grafted onto appropriate framework regions in the variable domains. Antigen-binding sites may be wild-type or modified by one or more amino acid substitutions, e.g. modified to resemble human immunoglobulins more closely. Some forms of humanized antibodies preserve all CDR sequences (for example a humanized mouse antibody which contains all six CDRs from the mouse antibody). Other forms have one or more CDRs which are altered with respect to the original antibody.
Different methods for humanizing antibodies are known to the skilled person, as reviewed by Almagro & Fransson, 2008, Frontiers in Bioscience, 13: 1619-1633, the content of which is herein incorporated by reference in its entirety. The review article by Almagro & Fransson is briefly summarized in US 2012/0231008 Al which is the national stage entry of international patent application WO 2011/063980 Al. The contents of US 2012/0231008 Al and WO 2011/063980 Al are herein incorporated by reference in their entirety.
As used herein, “human antibodies” include antibodies having variable and constant regions derived from human germline immunoglobulin sequences. The human antibodies of the invention may include amino acid residues not encoded by human germline immunoglobulin sequences (e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo). Human antibodies of the invention include antibodies isolated from human immunoglobulin libraries or from animals transgenic for one or more human immunoglobulin and that do not express endogenous immunoglobulins, as described for example in U.S. Patent No. 5,939,598 by Kucherlapati & Jakobovits.
The term “monoclonal antibody” as used herein refers to a preparation of antibody molecules of single molecular composition. A monoclonal antibody displays a single binding specificity and affinity for a particular epitope. In one embodiment, the monoclonal antibodies
are produced by a hybridoma which includes a B cell obtained from a non-human animal, e.g. mouse, fused to an immortalized cell.
The term “recombinant antibody”, as used herein, includes all antibodies that are prepared, expressed, created or isolated by recombinant means, such as (a) antibodies isolated from an animal (e.g., a mouse) that is transgenic or transchromosomal with respect to the immunoglobulin genes or a hybridoma prepared therefrom, (b) antibodies isolated from a host cell transformed to express the antibody, e.g. from a transfectoma, (c) antibodies isolated from a recombinant, combinatorial antibody library, and (d) antibodies prepared, expressed, created or isolated by any other means that involve splicing of immunoglobulin gene sequences to other DNA sequences.
The term “transfectoma”, as used herein, includes recombinant eukaryotic host cells expressing an antibody, such as CHO cells, NS/0 cells, HEK293 cells, HEK293T cells, plant cells, or fungi, including yeast cells.
As used herein, a “heterologous antibody” is defined in relation to a transgenic organism producing such an antibody. This term refers to an antibody having an amino acid sequence or an encoding nucleic acid sequence corresponding to that found in an organism not consisting of the transgenic organism, and being generally derived from a species other than the transgenic organism.
As used herein, a “heterohybrid antibody” refers to an antibody having light and heavy chains of different organismal origins. For example, an antibody having a human heavy chain associated with a murine light chain is a heterohybrid antibody.
Thus, “antibodies and antigen-binding fragments thereof’ suitable for use in the present invention include, but are not limited to, polyclonal, monoclonal, monovalent, bispecific, heteroconjugate, multispecific, recombinant, heterologous, heterohybrid, chimeric, humanized (in particular CDR-grafted), deimmunized, or human antibodies, Fab fragments, Fab' fragments, F(ab')2 fragments, fragments produced by a Fab expression library, Fd, Fv, disulfide- linked Fvs (dsFv), single chain antibodies (e.g. scFv), diabodies or tetrabodies (Holliger P. et al. (1993) Proc. Natl. Acad. Sci. U.S.A. 90(14), 6444-6448), nanobodies (also known as single domain antibodies), anti-idiotypic (anti-Id) antibodies (including, e.g., anti-Id antibodies to antibodies described herein), and epitope-binding fragments of any of the above.
The antibodies described herein are preferably isolated. An “isolated antibody” as used herein, is intended to refer to an antibody which is substantially free of other antibodies having different antigenic specificities (e.g., an isolated antibody that specifically binds to C5a is substantially free of antibodies that specifically bind antigens other than C5a). An isolated
antibody that specifically binds to an epitope, isoform or variant of human C5a may, however, have cross-reactivity to other related antigens, e.g. from other species (e.g. C5a species homologs, such as rat C5a). Moreover, an isolated antibody may be substantially free of other cellular material and/or chemicals. In one embodiment of the invention, a combination of “isolated” monoclonal antibodies relates to antibodies having different specificities and being combined in a well-defined composition.
The term “naturally occurring”, as used herein, as applied to an object refers to the fact that an object can be found in nature. For example, a polypeptide or polynucleotide sequence that is present in an organism (including viruses) that can be isolated from a source in nature and which has not been intentionally modified by man in the laboratory is naturally occurring.
Vilobelimab (sometimes referred to as IFX-1; InflaRx GmbH, Germany) is an antibody specifically binding to C5a. The CDR sequences and FR sequences of IFX-1 are disclosed in WO 2011/063980 Al (in Table 4 on page 51), the content of which is hereby incorporated by reference in its entirety. The affinity (Ka) and blocking activity is disclosed in WO 2011/063980 Al (in Table 3 on pages 49-50), the content of which is hereby incorporated by reference in its entirety.
BNJ364, BNJ367, BNJ371, BNJ378, BNJ366, BNJ369, BNJ381 and BNJ383 are antibodies specifically binding to C5a. The CDR sequences and FR sequences of these antibodies are disclosed in WO 2011/137395 Al (in Table 2 starting on page 55), the content of which is incorporated by reference in its entirety.
MEDI-7814 (Medlmmune) is a recombinant humanized anti-C5a antibody. The crystal structure of the human C5a in complex with MEDI7814 is available in the RCSB Protein Data Bank under 4UU9 (DOI: 10.2210/pdb4uu9/pdb).
ALXN-1007 (Alexion) is a humanized anti-C5a antibody.
Tocilizumab is a monoclonal antibody specifically binding to the IL-6 receptor (IL-6R). Tocilizumab (brand name Actemra) binds to soluble as well as membrane bound IL-6 receptor (sIL-6R and mIL-6R respectively). Tocilizumab is effective in blocking downstream IL-6 signalling.
Sarilumab (brand name Kevzara) is a further antibody binding to IL-6R that is suitable to exercise the invention. Sarilumab is a human monoclonal antibody binding to the IL-6 receptor.
Levilimab is a fully human monoclonal antibody acting against the interleukin-6 receptor (IL-6R), developed by Biocad, for the treatment of rheumatoid arthritis. Levilimab targets membrane bound and soluble IL-6 receptor.
Vobarilizumab (synonym ALX-0061) is an high affinity anti-IL-6R bispecific nanobody further targeting albumin to extend half-life.
Siltuximab (brand name Sylvant) is a chimeric monoclonal antibody that binds to interleukin-6 (IL-6), preventing binding to soluble and membrane bound interleukin-6 receptors. Siltuximab is FDA-approved for treatment of patients with idiopathic multicentric Castleman’s disease (iMCD) who do not have human immunodeficiency virus (HIV) or human herpesvirus-8 (HHV-8).
Sirukumab is a human monoclonal antibody specifically binding to IL-6. Sirukumab is under development for the treatment of rheumatoid arthritis.
Olokizumab is a humanized monoclonal antibody specifically binding to IL-6. Olokizumab has been tested for treatment of rheumatoid arthritis and has been used as an emergency experimental cytokine storm COVID-19 treatment.
Elsilimomab is a monoclonal antibody that targets and blocks IL-6. Elsilimomab has been under development for treatment of lymphoma and myeloma.
Clazakizumab is a humanized monoclonal antibody directed against IL-6. Clazakizumab has been under development for treatment of psoriatic arthritis.
PF-4236921 (sometimes referred to as PF-04236921) is an anti-IL-6 monoclonal antibody that has been under development for the treatment of rheumatoid arthritis, systemic lupus erythematosus and Crohn's disease.
EBL031 (sometimes referred to as RG-6179) is a humanised monoclonal antibody that specifically binds to IL-6.
As used herein, a “patient” means any mammal or bird who may benefit from a treatment with the compound described herein (i.e. with an inhibitor of C5a activity described herein). Preferably, a “patient” is selected from the group consisting of laboratory animals (e.g. mouse or rat), domestic animals (including e.g. guinea pig, rabbit, chicken, turkey, pig, sheep, goat, camel, cow, horse, donkey, cat, or dog), or primates including chimpanzees and human beings. It is particularly preferred that the “patient” is a human being.
As used herein, "treat", "treating" or “treatment” of a disease or disorder means accomplishing one or more of the following: (a) reducing the severity and/or duration of the disorder; (b) limiting or preventing development of symptoms characteristic of the disorder(s) being treated; (c) inhibiting worsening of symptoms characteristic of the disorder(s) being treated; (d) limiting or preventing recurrence of the disorder(s) in patients that have previously had the disorder(s); and (e) limiting or preventing recurrence of symptoms in patients that were previously symptomatic for the disorder(s).
As used herein, “prevent”, “preventing”, “prevention”, or “prophylaxis” of a disease or disorder means preventing that a disorder occurs in subject.
An “effective amount” is an amount of a therapeutic agent sufficient to achieve the intended purpose. The effective amount of a given therapeutic agent will vary with factors such as the nature of the agent, the route of administration, the size and species of the animal to receive the therapeutic agent, and the purpose of the administration. The effective amount in each individual case may be determined empirically by a skilled artisan according to established methods in the art.
The term “active agent” as used herein, refers to any therapeutic activity an agent may exhibit.
“Pharmaceutically acceptable” means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans.
The term “carrier”, as used herein, refers to a diluent, adjuvant, excipient, or vehicle with which the therapeutic agent is administered. Such pharmaceutical carriers can be sterile liquids, such as saline solutions in water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. A saline solution is a preferred carrier when the pharmaceutical composition is administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions. Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like. The composition, if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents. These compositions can take the form of solutions, suspensions, emulsions, tablets, pills, capsules, powders, sustained-release formulations and the like. The composition can be formulated as a suppository, with traditional binders and carriers such as triglycerides. The compounds of the invention can be formulated as neutral or salt forms. Pharmaceutically acceptable salts include those formed with free amino groups such as those derived from hydrochloric, phosphoric, acetic, oxalic, tartaric acids, etc., and those formed with free carboxyl groups such as those derived from sodium, potassium, ammonium, calcium, ferric hydroxides, isopropylamine, triethylamine, 2-ethylamino ethanol, histidine, procaine, etc. Examples of suitable pharmaceutical carriers are described in "Remington's Pharmaceutical Sciences" by E. W. Martin. Such compositions will contain a therapeutically effective amount of the compound,
preferably in purified form, together with a suitable amount of carrier so as to provide the form for proper administration to the patient. The formulation should suit the mode of administration.
Embodiments
In the following different aspects of the invention are defined in more detail. Each aspect so defined may be combined with any other aspect or aspects unless clearly indicated to the contrary. In particular, any feature indicated as being preferred or advantageous may be combined with any other feature or features indicated as being preferred or advantageous.
In a first aspect, the present invention provides an inhibitor of C5a activity for use in:
(i) combination with an inhibitor of IL-6 activity in the treatment of infectious pneumonia and/or infectious acute respiratory distress syndrome (ARDS); and/or
(ii) the treatment of infectious pneumonia and/or infectious acute respiratory distress syndrome (ARDS), wherein the patient is or has been treated with an inhibitor of IL-6 activity.
In combination treatment the inhibitor of C5a activity and the inhibitor of IL-6 activity are preferably administered within the half-life of the other inhibitor. For example, if an inhibitor of C5a activity with a half-life of about 4 days and an inhibitor of IL-6 activity with a half-life of about 13 days is used, then the inhibitor of IL-6 activity would be administered within about 3 days after the administration of the inhibitor of C5a activity or if the inhibitor of IL-6 activity is administered first, the inhibitor of C5a activity is administered within 13 days after the inhibitor of IL-6 activity. Thus an overlap between the action of both inhibitors should exist in combination treatment.
In a preferred embodiment the inhibitor of C5a activity and the inhibitor of IL-6 activity are preferably administered within 1-5 (i.e. 1, 2, 3, 4 or 5) half-lives of the other inhibitor.
In a preferred embodiment of the first aspect of the invention the inhibitor of C5a activity and the inhibitor of IL-6 activity are administered on the same day.
In a preferred embodiment of the first aspect of the invention the inhibitor of IL-6 activity is administered within 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 days prior or after the administration of the inhibitor of C5a activity.
In a preferred embodiment of the first aspect of the invention the inhibitor of C5a activity is administered within 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 days prior or after the administration of the inhibitor of IL-6 activity.
The patients benefiting most from the use of the inhibitor of C5a activity in the treatment of infectious pneumonia and/or infectious acute respiratory distress syndrome (ARDS) are patients that are or have been treated with an inhibitor of IL-6 activity. In a preferred
embodiment the inhibitor of IL-6 activity is or has been administered to the patient within a time-window so that there is an overlap between the action of the inhibitor of IL-6 activity and the inhibitor of C5a activity, at least with regard to the administration of the first dose of the inhibitor of C5a activity.
In a second aspect, the present invention provides an inhibitor of IL-6 activity for use in:
(i) combination with an inhibitor of C5a activity in the treatment of infectious pneumonia and/or infectious acute respiratory distress syndrome (ARDS); and/or
(ii) the treatment of infectious pneumonia and/or infectious acute respiratory distress syndrome (ARDS), wherein the patient is or has been treated with an inhibitor of C5a activity.
In combination treatment the inhibitor of IL-6 activity and the inhibitor of C5a activity are preferably administered within the half-life of the other inhibitor. For example, if an inhibitor of IL-6 activity with a half-life of about 13 days and an inhibitor of C5a activity with a half-life of about 4 days is used, then the inhibitor of IL-6 activity would be administered within about 4 days after the administration of the inhibitor of C5a activity or if the inhibitor of IL-6 activity is administered first, the inhibitor of C5a activity is administered within 13 days after the inhibitor of IL-6 activity. Thus an overlap between the action of both inhibitors should exist in combination treatment.
In a preferred embodiment of the first aspect of the invention the inhibitor of IL-6 activity and the inhibitor of C5a activity are administered on the same day.
In a preferred embodiment of the first aspect of the invention the inhibitor of IL-6 activity is administered within 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 days after the administration of the inhibitor of C5a activity.
In a preferred embodiment of the first aspect of the invention the inhibitor of C5a activity is administered within 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 days after the administration of the inhibitor of IL-6 activity.
The patients benefiting most from the use of the inhibitor of IL-6 activity in the treatment of infectious pneumonia and/or infectious acute respiratory distress syndrome (ARDS) are patients that are or have been treated with an inhibitor of C5a activity. In a preferred embodiment the inhibitor of C5a activity is or has been administered to the patient within a time-window so that there is an overlap between the action of the inhibitor of IL-6 activity and the inhibitor of C5a activity, at least with regard to the administration of the first dose of the inhibitor of IL-6 activity.
Inhibitors of IL-6 activity
In a preferred embodiment of all aspects of the present invention, the inhibitor of IL-6 activity specifically binds to IL-6 or the IL-6 receptor. In a preferred embodiment the inhibitor of IL-6 activity comprises a protein ligand. In a preferred embodiment the inhibitor of IL-6 activity is a protein ligand. In a preferred embodiment the inhibitor of IL-6 activity is an antibody or antigen binding fragment thereof specifically binding to the IL-6 receptor. In a preferred embodiment the inhibitor of IL-6 activity is an antibody or antigen binding fragment thereof specifically binding to IL-6.
In a preferred embodiment according to all aspects of the invention the inhibitor of IL- 6 activity is specifically binding to IL-6R, preferably selected from Tocilizumab, Sarilumab, Levilimab and Vobarilizumab. Preferably the inhibitor of IL-6R is Tocilizumab.
In a preferred embodiment according to all aspects of the invention the inhibitor of IL- 6 activity is specifically binding to IL-6R, preferably selected from Siltuximab, Sirukumab, Olokizumab, Elsilimomab, Clazakinumab, PF-423691 and EBI-031.
Inhibitors of C5a activity
In a preferred embodiment of all aspects of the invention, the inhibitor of C5a activity binds specifically to C5a or to the C5a receptor (C5aR). In a preferred embodiment, the inhibitor of C5a activity binds specifically to C5a. In a preferred embodiment, the inhibitor of C5a activity binds specifically to C5aR. In a preferred embodiment the inhibitor of C5a activity comprises a protein ligand specifically binding to C5a or the C5aR. In a preferred embodiment the inhibitor of C5a activity comprises a protein ligand specifically binding to C5a. In a preferred embodiment the inhibitor of C5a activity comprises a protein ligand specifically binding to C5aR.
In a preferred embodiment the inhibitor of C5a activity, is a small molecule C5aR antagonist. Suitable small molecule C5aR antagonists are known in the art and can inter alia be found in WO 2020/182384 Al, which is incorporated by reference herein. Methods for preparing the small molecule C5aR antagonists can be found in WO 2020/182384 AL
In a preferred embodiment the inhibitor of C5a activity is a small molecule having the general formula (I)
and pharmaceutically acceptable salts, hydrates and rotamers thereof; wherein
C1 is selected from the group consisting of aryl and heteroaryl, wherein the heteroaryl group has from 1-3 heteroatoms as ring members selected from N, O and S; and wherein said aryl and heteroaryl groups are optionally substituted with from 1 to 3 R1 substituents;
C2 is selected from the group consisting of aryl and heteroaryl, wherein the heteroaryl group has from 1-3 heteroatoms as ring members selected from N, O and S; and wherein said aryl and heteroaryl groups are optionally substituted with from 1 to 3 R2 substituents;
C3 is selected from the group consisting of Ci-8 alkyl or heteroalkyl, C3-8 cycloalkyl, C3-8 cycloalkyl-Ci-4 alkyl, aryl, aryl-Ci-4 alkyl, heteroaryl, heteroaryl-Ci-4 alkyl, heterocycloalkyl or heterocycloalkyl-Ci-4 alkyl, wherein the heteroalkyl group has from 1-3 heteroatoms selected from N, O and S, wherein the heterocycloalkyl group or portion has from 1-3 heteroatoms selected from N, O and S, and wherein the heteroaryl group has from 1-3 heteroatoms as ring members selected from N, O and S, and each C3 is optionally substituted with from 1 to 3 R3 substituents; each R1 is independently selected from the group consisting of halogen, — CN, -Rc, — CO2Ra, — CONRaRb, — C(O)Ra, — OC(O)NRaRb, — NRbC(O)Ra, — NRbC(O)2Rc, — NRa— C(O)NRaRb, — NRaC(O)NRaRb, — NRaRb, — ORa, and — S(O)2NRaRb; wherein each Ra and Rb is independently selected from hydrogen, C1-8 alkyl, and C1-8 haloalkyl, or when attached to the same nitrogen atom can be combined with the nitrogen atom to form a five or six-membered ring having from 0 to 2 additional heteroatoms as ring members selected from N, O or S, and is optionally substituted with one or two oxo; each Rc is independently selected from the group consisting of C1-8 alkyl or heteroalkyl, C1-8 haloalkyl, C3-6 cycloalkyl, heterocycloalkyl, aryl and heteroaryl, and wherein the aliphatic and/or cyclic portions of Ra, Rb and Rc are optionally further substituted with from one to three halogen, hydroxy, methyl, amino, alkylamino and dialkylamino groups; and optionally when two R1 substituents are on adjacent atoms, are combined to form a fused five or six-membered carbocyclic or heterocyclic ring;
each R2 is independently selected from the group consisting of halogen, — CN, — NO2, — Rf, — CO2Rd, — CONRdRe, — C(O)Rd, — OC(O)NRdRe, — NReC(O)Rd, — NReC(O)2Rf, — NRdC(O)NRdRe, — NRdRe, — ORd, and — S(O)2NRdRe; wherein each Rd and Re is independently selected from hydrogen, C1-8 alkyl, and C1-8 haloalkyl, or when attached to the same nitrogen atom can be combined with the nitrogen atom to form a five or sixmembered ring having from 0 to 2 additional heteroatoms as ring members selected from N, O or S, and is optionally substituted with one or two oxo; each Rf is independently selected from the group consisting of C1-8 alkyl or heteroalkyl, C1-8 haloalkyl, C3-6 cycloalkyl, heterocycloalkyl, aryl and heteroaryl, and wherein the aliphatic and/or cyclic portions of Rd, Re and Rfare optionally further substituted with from one to three halogen, hydroxy, methyl, amino, alkylamino and dialkylamino groups, and optionally when two R2 groups are on adjacent atoms, they are combined to form a five- or six-membered ring; each R3 is independently selected from the group consisting of halogen, — CN, — R1, — CO2Rg, — CONRgRh, — C(O)Rg, — C(O)R', — OC(O)NRgRh, — NRhC(O)Rg, — NRhCO2Ri, — NRSQOjNRSR11, — NRgRh, — ORg, — ORj, S(O)2NRgRh, — X4— Rj, — NH— X4— R>,
— O— X4— Rj, — X4— NRgRh, — X4— NHR>, — X4— CONRgRh, — X4— NRhC(O)Rg, — X4— CO2Rg, — O— X4— CO2Rg, — NH— X4— CO2Rg, — X4— NRhCO2Ri, — O— X4 — NR11CO2RI, — NHR1 and — NHCH2R1, wherein X4 is a C1-4 alkylene; each Rg and Rh is independently selected from hydrogen, C1-8 alkyl or heteroalkyl, C3-6 cycloalkyl and C1-8 haloalkyl, or when attached to the same nitrogen atom can be combined with the nitrogen atom to form a four-, five- or six-membered ring having from 0 to 2 additional heteroatoms as ring members selected from N, O or S and is optionally substituted with one or two oxo; each R1 is independently selected from the group consisting of C1-8 alkyl or heteroalkyl, C1-8 haloalkyl, C3-6 cycloalkyl, heterocycloalkyl, aryl and heteroaryl; and each R> is selected from the group consisting of C3-6 cycloalkyl, imidazolyl, pyrimidinyl, pyrrolinyl, pyrrolyl, piperidinyl, morpholinyl, tetrahydrofuranyl, tetrahydropyranyl, and S,S-dioxo-tetrahydrothiopyranyl, and wherein the aliphatic and/or cyclic portions of Rg, Rh, R1 and R1 are optionally further substituted with from one to three halogen, methyl, CF3, hydroxy, C1-4 alkoxy, C1-4 alkoxy-Ci-4 alkyl, — C(O)O — C1-8 alkyl, amino, alkylamino and dialkylamino groups, and optionally when two R3 groups are on adjacent atoms, they are combined to form a five- or six-membered ring;
X is hydrogen or CH3; and
R8 and R9 are independently from each other selected from the group consisting of hydrogen, halogen, Ci-Cs alkyl, Ci-Cs haloalkyl, and Ci-Cs alkoxy or R8 and R9 are combined to form a fused saturated or unsaturated mono- or multi-ring carbocycle in which one or more of the ring carbon atoms may be replaced independently from each other by N, S, or O, with the proviso that at least one of R8 and R9 is not hydrogen.
In a preferred embodiment the inhibitor of C5a activity is a small molecule C5aR antagonist having the general formula (I) as indicated above and wherein R8 and/or R9 is hydrogen.
In a preferred embodiment the inhibitor of C5a activity is selected from:
INF 004 : (2R,3 S)-2-(4-(cyclopentylamino)phenyl)- 1 -(2-fluoro-6-methylbenzoyl)-
N-(4-methyl-3-(trifluoromethyl)phenyl)-l,2,3,4-tetrahydroquinoline-3-carboxamide
INF011 : (2R,3S)-l-(2-fluoro-6-methylbenzoyl)-N-(4-methyl-3-
(trifluoromethyl)phenyl)-2-(4-((tetrahydro-2H-pyran-4-yl)amino)phenyl)-l,2,3,4- tetrahydroquinoline-3-carboxamide
INFO 14: (2R,3S)-l-(2-chlorobenzoyl)-2-(4-(cyclopentylamino)phenyl)-N-(4-
(hydroxymethyl)-3 -(tri fluoromethyl Jphenyl)- 1,2, 3, 4-tetrahydroquinoline-3 -carboxamide
INFO 15 : (2R,3 S)-2-(4-(cyclopentylamino)phenyl)- 1 -(2-fluoro-6-methylbenzoyl)-
N-(4-(hydroxymethyl)-3 -(tri fluoromethyl (phenyl)- 1,2, 3, 4-tetrahydroquinoline-3 -carboxamide
INF022: (2R,3S)-2-(4-(cyclopentylamino)phenyl)-6-fluoro-l-(2-fluoro-6- methylbenzoyl)-N-(4-methyl-3-(tri fluoromethyl)phenyl)- 1,2,3, 4-tetrahydroquinoline-3- carb oxami de
INF023: (2R,3S)-2-(4-(cyclopentylamino)phenyl)-l-(2-fluoro-6-methylbenzoyl)-
6-methyl-N-(4-methyl-3 -(tri fluoromethyl)phenyl)- 1,2, 3, 4-tetrahydroquinoline-3 -carboxamide
INF024: (2R,3S)-2-(4-(cyclopentylamino)phenyl)-l-(2-fluoro-6-methylbenzoyl)-
6-methoxy-N-(4-methyl-3 -(tri fluoromethyl)phenyl)- 1,2,3, 4-tetrahydroquinoline-3 - carboxamide
INF025: (2R,3S)-2-(4-(cyclopentylamino)phenyl)-6,7-difluoro-l-(2-fluoro-6- methylbenzoyl)-N-(4-methyl-3-(tri fluoromethyl)phenyl)- 1,2,3, 4-tetrahydroquinoline-3- carb oxami de
INF030 : (2R, 3 S)- 1 -(2-fluoro-6-methylbenzoyl)-N-(4-(hydroxymethyl)-3 -
(trifluoromethyl)phenyl)-2-(4-((tetrahydro-2H-pyran-4-yl)amino)phenyl)-l,2,3,4- tetrahydroquinoline-3-carboxamide
INF033: (2R,3S)-2-(4-(cyclopentylamino)phenyl)-l-(2-fluoro-6-methylbenzoyl)-
N-(4-hydroxy-3-(trifluoromethyl)phenyl)-l,2,3,4-tetrahydroquinoline-3-carboxamide
INF034 : (2R, 3 S)- 1 -(2-fluoro-6-methylbenzoyl)-2-(4-(3 -hydroxy-3 - methylbutyl)phenyl)-N-(4-methyl-3-(trifluoromethyl)phenyl)-l,2,3,4-tetrahydroquinoline-3- carb oxami de
INF035: (2R,3S)-2-(4-(cyclopentylamino)phenyl)-l-(2-fluoro-6-methylbenzoyl)-
N-(3-(hydroxymethyl)phenyl)-l,2,3,4-tetrahydroquinoline-3-carboxamide
INF038: (2R,3S)-l-(2-fluoro-6-methylbenzoyl)-N-(4-(hydroxymethyl)-3-
(trifluoromethyl)phenyl)-2-phenyl-l,2,3,4-tetrahydroquinoline-3-carboxamide
INF039: (2R,3S)-2-(4-(cyclopentylamino)phenyl)-6-fluoro-l-(2-fluoro-6- methylbenzoyl)-N-(4-(hydroxymethyl)-3-(trifluoromethyl)phenyl)-l,2,3,4- tetrahydroquinoline-3-carboxamide
INF040: (2R,3S)-2-(4-(cyclopentylamino)phenyl)-N-(4-(hydroxymethyl)-3-
(trifluoromethyl)phenyl)-l-(2-methylbenzoyl)- 1,2, 3, 4-tetrahydroquinoline-3 -carboxamide
INF041: (2R,3S)-2-(4-(cyclopentylamino)phenyl)-l-(2-fluorobenzoyl)-N-(4-
(hydroxymethyl)-3-(trifluoromethyl)phenyl)- 1,2, 3, 4-tetrahydroquinoline-3 -carboxamide
INF045: (2R,3S)-2-(4-(cyclopentylamino)phenyl)-5-fluoro-l-(2-fluoro-6- methylbenzoyl)-N-(4-(hydroxymethyl)-3-(trifluoromethyl)phenyl)-l,2,3,4- tetrahydroquinoline-3-carboxamide
INF046: (2R,3S)-2-(4-(cyclopentylamino)phenyl)-7-fluoro-l-(2-fluoro-6- methylbenzoyl)-N-(4-(hydroxymethyl)-3-(trifluoromethyl)phenyl)-l,2,3,4- tetrahydroquinoline-3-carboxamide
INF047: (2R,3S)-6-chloro-2-(4-(cyclopentylamino)phenyl)-l-(2-fluoro-6- methylbenzoyl)-N-(4-(hydroxymethyl)-3-(trifluoromethyl)phenyl)-l,2,3,4- tetrahydroquinoline-3-carboxamide
INF048: (2R,3S)-l-(2-fluoro-6-methylbenzoyl)-2-(4-(3-hydroxy-3- methylbutyl)phenyl)-N-(4-(hydroxymethyl)-3-(trifluoromethyl)phenyl)-l,2,3,4- tetrahydroquinoline-3-carboxamide
INF049: (2R,3S)-l-(2-fluoro-6-methylbenzoyl)-N-(4-(hydroxymethyl)-3-
(trifluoromethyl)phenyl)-2-(4-(isopropylamino)phenyl)-l,2,3,4-tetrahydroquinoline-3- carb oxami de
INF050: (2R,3S)-2-(4-(cyclobutylamino)phenyl)-l-(2-fluoro-6-methylbenzoyl)-
N-(4-(hydroxymethyl)-3-(trifluoromethyl)phenyl)- 1,2, 3, 4-tetrahydroquinoline-3 -carboxamide
INF051: (2R,3S)-l-(2-fluoro-6-methylbenzoyl)-2-(4-((2-hydroxy-2- methylpropyl)amino)phenyl)-N-(4-(hydroxymethyl)-3-(trifluoromethyl)phenyl)-l,2,3,4- tetrahydroquinoline-3-carboxamide
INF052: (2R,3S,5R)-2-(4-(cyclopentylamino)phenyl)-l-(2,6-difluorobenzoyl)-
N-(4-methyl-3-(trifluoromethyl)phenyl)-5-(trifluoromethyl)piperidine-3-carboxamide
INF053: (2R,3S,5R)-2-(4-(cyclopentylamino)phenyl)-l-(2,6-dimethylbenzoyl)-
N-(4-methyl-3-(trifluoromethyl)phenyl)-5-(trifluoromethyl)piperidine-3-carboxamide
INF055: (2R,3S)-2-(4-(cyclopentylamino)phenyl)-l-(2-fluoro-6-methylbenzoyl)-
N-(4-(hydroxymethyl)-3-(trifluoromethyl)phenyl)-6-(trifluoromethyl)-l,2,3,4- tetrahydroquinoline-3-carboxamide
INF056: (2R,3S,5R)-2-(4-(cyclopentylamino)phenyl)-l-(2-fluoro-6- methylbenzoyl)-N-(4-methyl-3-(trifluoromethyl)phenyl)-5-(trifluoromethyl)piperidine-3- carb oxami de
INF058 : (2R, 3 S)- 1 -(2-fluoro-6-methylbenzoyl)-2-(4-(2-hydroxy-2- methylpropoxy)phenyl)-N-(4-methyl-3-(trifluoromethyl)phenyl)- 1,2,3, 4-tetrahydroquinoline- 3 -carboxamide
INF067: (2R,3S,5R)-2-(4-((cyclopentyl-l-d)amino)phenyl)-l-(2,6- difluorobenzoyl)-N-(4-methyl-3-(trifluoromethyl)phenyl)-5-(trifluoromethyl)piperidine-3- carboxamide
INF068: (2R,3S,5R)-2-(4-(cyclopentylamino)phenyl)-l-(2,6-difluorobenzoyl)-
N-(4-hydroxy-3-(trifluoromethyl)phenyl)-5-(trifluoromethyl)piperidine-3-carboxamide
INF069: (2R,3S,5R)-2-(4-(cyclopentylamino)phenyl)-l-(2,6-difluorobenzoyl)-
N-(4-(hydroxymethyl)-3-(trifluoromethyl)phenyl)-5-(trifluoromethyl)piperidine-3- carboxamide
INF070: (2R,3S,5R)-l-(2,6-difluorobenzoyl)-N-(4-methyl-3-
(trifluoromethyl)phenyl)-2-(4-((tetrahydro-2H-pyran-4-yl)amino)phenyl)-5- (trifluoromethyl)piperidine-3-carboxamide
INF071 : (2R,3S,5R)-N-(3-chloro-4-methylphenyl)-2-(4-
(cyclopentylamino)phenyl)-l-(2,6-difluorobenzoyl)-5-(trifluoromethyl)piperidine-3- carb oxami de
INF072: (2R,3S,5R)-N-(3-chloro-4-hydroxyphenyl)-2-(4-
(cyclopentylamino)phenyl)-l-(2,6-difluorobenzoyl)-5-(trifluoromethyl)piperidine-3- carb oxami de
INF075: (2R,3S,5R)-l-(2-chloro-6-fluorobenzoyl)-2-(4-
(cyclopentylamino)phenyl)-N-(4-methyl-3-(trifluoromethyl)phenyl)-5-
(trifluoromethyl)piperidine-3-carboxamide
INF077: (2R,3S,5R)-N-(4-chloro-3-(trifluoromethyl)phenyl)-2-(4-
(cyclopentylamino)phenyl)-l-(2,6-difluorobenzoyl)-5-(trifluoromethyl)piperidine-3- carb oxami de
INF080: (2R,3S,5R)-2-(4-(cyclopentylamino)phenyl)-N-(3,4-dichlorophenyl)-l-
(2,6-difluorobenzoyl)-5-(trifluoromethyl)piperidine-3-carboxamide
Methods for preparing these small molecule C5aR antagonists can be found in WO 2020/182384 Al.
In a preferred embodiment the inhibitor of C5a activity is INF052.
In a preferred embodiment the inhibitor of C5a activity is avacopan, having the structure of general formula (II):
In a preferred embodiment of all aspects of the present invention, the inhibitor of C5a activity is a protein ligand, wherein the protein ligand is an antibody or an antigen binding fragment thereof. More preferably the protein ligand comprises a VH and a VL domain of an antibody, a Fab fragment, a Fab’ fragment, a heavy chain antibody, a single-domain antibody (sdAb), variable domain of a heavy chain antibody, a VHH, a nanobody, a single-chain variable fragment (scFv), a tandem scFv, or a single-chain diabody.
In a preferred embodiment of all aspects of the present invention, the inhibitor of C5a activity is a protein ligand, wherein the protein ligand is an antibody-like protein, preferably an affibody, anticalin, or an designed ankyrin repeat protein. Antibody-like proteins include without limitation affibodies, anticalins, and designed ankyrin repeat proteins (for review see: Binz H.K. et al. (2005) Engineering novel binding proteins from non-immunoglobulin domains. Nat. Biotechnol. 23(10): 1257-1268). Antibody-like proteins can be derived from large libraries of mutants, e.g. be panned from large phage display libraries and can be isolated in analogy to regular antibodies. Also, antibody-like binding proteins can be obtained by combinatorial
mutagenesis of surface-exposed residues in globular proteins. Antibody-like proteins are sometimes referred to as “peptide aptamers”.
In a preferred embodiment of all aspects of the present invention, the inhibitor of C5a activity is a protein ligand, wherein the protein ligand is an inhibitory variant of C5a.
In a preferred embodiment of all aspects of the present invention, the inhibitor of C5a activity is a protein ligand, wherein the protein ligand is an inhibitory variant of C5a receptor.
In preferred embodiments of all aspects of the invention, wherein the inhibitor of C5a activity is a protein ligand, preferably an antibody or antigen-binding fragment thereof, specifically binding to a conformational epitope of human C5a formed by (a) amino acid sequences NDETCEQRA (SEQ ID NO: 2) and SHKDMQL (SEQ ID NO: 3) of C5a binds to a conformational epitope of C5a formed by amino acid sequences NDETCEQRA (SEQ ID NO: 2) and SHKDMQL (SEQ ID NO: 3), and binds to at least one amino acid within the amino acid sequence according to SEQ ID NO: 2 and to at least one amino acid within the amino acid sequence according to SEQ ID NO: 3. In other words, the inhibitor of C5a activity according to this preferred embodiment binds at the same time to at least one amino acid within the amino acid sequence according to SEQ ID NO: 2 and to at least one amino acid within the amino acid sequence according to SEQ ID NO: 3. SEQ ID NO: 2 corresponds to amino acids 30-38 of human C5a. SEQ ID NO: 3 corresponds to amino acids 66-72 of human C5a. The amino acid sequence of human C5a is depicted in SEQ ID NO: 1. In more preferred embodiments, the inhibitor of C5a activity binds to at least one of amino acids DETCEQR (SEQ ID NO: 4). SEQ ID NO: 4 corresponds to amino acids 31-37 of human C5a. In more preferred embodiments, the inhibitor of C5a activity binds to at least one of amino acids HKDMQ (SEQ ID NO: 5), more preferably to at least one of amino acids KDM. SEQ ID NO: 5 correspond to amino acids 67-71 of human C5a; the sequence KDM corresponds to amino acids 68-70 of human C5a. In particularly preferred embodiments, the inhibitor of C5a activity binds at the same time to at least one amino acid within the amino acid sequence DETCEQR (SEQ ID NO: 4) and to at least one amino acid within the amino acid sequence KDM.
In a preferred embodiment the specific binding to the amino acids of the conformational epitope is determined by alanine scanning. In a preferred embodiment the conformational epitope is determined by alanine scanning.
The skilled person is aware of assays for screening antibodies binding to the specified conformational epitope. One such suitable assay is described in the following:
In 3-D structure of C5a obtained from computer modeling method, the spatial epitopes containing peptide C5a 28-40 (VNNDETCEQRAAR, SEQ ID NO: 53) and C5a peptide 65-70
(ISHKDM, SEQ ID NO: 54) can be viewed as random coils. When the two peptides are linked by a flexible peptide linker, GGGGS (SEQ ID NO: 55), the spatial epitopes is reconstructed resembling the parent antigen conformation, as the weak hydrophobic interaction from the two peptides ensures a pocket-shape conformation. Computer modeling analysis of the peptide NH2-28-40-Linker(GGGGS)-65-70-COOH maintains the same conformation as the parent antigen. This new 24-AA peptide can be synthesized and conjugated with keyhole limpet hemocyanin (KLH) to form an immunogen to immunize mice, and the traditional hybridoma technology can be subsequently applied to obtain INab308 and INab708 using the new 24-AA peptide based ELISA as a screening tool. A 96-well ELISA plate is coated with 1-2 wg/mL synthetic peptides with the conformational epitope at 4°C overnight. After being blocked with 5% nonfat milk in PBS at 37°C for 1 h, 50 «L of culture media of the hybridoma growing clones are added to each well and incubated at 37°C for 1 h, followed by 100 «L of goat antimouse antibody labeled with horseradish peroxidase (HRP) for 1 h. The peroxidase reaction is developed with color development solution containing 5.5 mM o-phenylene-diamine hydrochloride (OPD) and 8.5 mM H2O2. The light absorbance is measured at 492 nm with an ELISA reader.
In a preferred embodiment of all aspects of the present invention wherein the inhibitor of C5a activity is an antibody or antigen-binding fragment thereof specifically binding to C5a comprising a variable domain of a heavy chain (VH) and a variable domain of a light chain (VL).
Preferably the VL comprises a light chain CDR3 sequence CQQSNEDPYT as set forth in SEQ ID NO: 6. In a preferred embodiment the VL further comprises a FR2 sequence WYQQKPGQPPKLL as set forth in SEQ ID NO: 8 or SEQ ID NO: 39.
Preferably the VL comprises a light chain CDR3 sequence CQQNNEDPLT as set forth in SEQ ID NO: 7. In a preferred embodiment the VL further comprises a FR2 sequence WYQQKPGQPPKLL as set forth in SEQ ID NO: 8 or SEQ ID NO: 39.
In a preferred embodiment the inhibitor of C5a activity further comprises at least one of the following sequences:
(i) a heavy chain CDR2 sequence IDPSDSESRLDQ according to SEQ ID NO: 9;
(ii) a heavy chain CDR2 sequence ILPGSGSTNYNE according to SEQ ID NO: 10;
(iii) a light chain CDR2 sequence IYAASNL according to SEQ ID NO: 11;
(iv) a light chain CDR2 sequence IYAASNL according to SEQ ID NO: 12;
(v) a heavy chain CDR1 sequence CKASGYSFTTFWMD according to SEQ ID NO: 13;
(vi) a heavy chain CDR1 sequence CKATGNTFSGYWIE according to SEQ ID NO: 14;
(vii) a light chain CDR1 sequence CKASQSVDYDGDSYMK according to SEQ ID NO: 15;
(viii) a light chain CDR1 sequence CKASQSVDYDGDSYMN according to SEQ ID NO: 16;
(ix) a heavy chain CDR3 sequence CARGNDGYYGFAY according to SEQ ID NO: 17; or
(x) a heavy chain CDR3 sequence CTRRGLYDGSSYFAY according to SEQ ID NO: 18.
In a preferred embodiment the inhibitor of C5a activity comprises a light chain CDR3 sequence according to SEQ ID NO: 6, a light chain CDR2 sequence according to SEQ ID NO:
11, a light chain CDR1 sequence according to SEQ ID NO: 15, a heavy chain CDR3 sequence according to SEQ ID NO: 17, a heavy chain CDR2 sequence according to SEQ ID NO: 9, and a heavy chain CDR1 sequence according to SEQ ID NO: 13. In a preferred embodiment the inhibitor of C5a activity further comprises the light chain framework regions (FR) 1 to 4 according to SEQ ID NOs: 19, 8, 20 and 21, respectively; preferably further comprising the heavy chain FR 1 to 4 according to SEQ ID NOs: 23, 24, 25 and 26, respectively. In an alternative preferred embodiment the inhibitor of C5a activity further comprises the light chain framework regions (FR) 1 to 4 according to SEQ ID NOs: 37, 39, 41 and 43, respectively; preferably further comprising the heavy chain FR 1 to 4 according to SEQ ID NOs:45, 47, 49 and 51, respectively.
In a preferred embodiment the inhibitor of C5a activity comprises a light chain CDR3 sequence according to SEQ ID NO: 7, a light chain CDR2 sequence according to SEQ ID NO:
12, and a light chain CDR1 sequence according to SEQ ID NO: 16, a heavy chain CDR3 sequence according to SEQ ID NO: 18, a heavy chain CDR2 sequence according to SEQ ID NO: 10, and a heavy chain CDR1 sequence according to SEQ ID NO: 14. In a preferred embodiment the inhibitor of C5a activity further comprises the light chain framework regions (FR) 1 to 4 according to SEQ ID NOs:28, 8, 29 and 30, respectively; preferably further comprising the heavy chain FR 1 to 4 according to SEQ ID NOs:32, 33, 34 and 35, respectively.
In a preferred embodiment the FR sequences have a sequence identity to the herein disclosed FR sequences of at least 99%, at least 98% at least 95%, at least 90%, at least 85%, at least 80% or at least 75% sequence.
In a preferred embodiment according to all ascpects of the invention the inhibitor of C5a activity is selected from IFX-1 (vilobelimab), IFX-2, INab708, BNJ364, BNJ367, BNJ371, BNJ378, BNJ366, BNJ369, BNJ381, BNJ383, MEDI-7814 or ALXN-1007, or an antigenbinding fragment thereof. In a preferred embodiment according to all ascpects of the invention the inhibitor of C5a activity is IFX-1 (vilobelimab).
Table 1: FR1, CDR1, FR2, CDR2, FR3, CDR3, and FR4 sequences of VL and VH domains of suitable inhibitors of C5a activity
In a preferred embodiment according to all ascpects of the invention the inhibitor of C5a activity is selected from an antibody or antigen binding fragment thereof that competes for binding with any of the antibodies selected from:IFX-l (vilobelimab), IFX-2, INab708, BNJ364, BNJ367, BNJ371, BNJ378, BNJ366, BNJ369, BNJ381, BNJ383, MEDI-7814 or ALXN-1007.
In a preferred embodiment according to all aspects of the invention the inhibitor of C5a activity is an antibody or antigen-binding fragment thereof that specifically binds to human C5a comprises (i) a light chain variable domain (VL) having at least 99%, at least 98% at least 95%, at least 90%, at least 85%, at least 80%, at least 75% sequence identity with the VL domain according to SEQ ID NO: 22 and (ii) a heavy chain variable domain (VH) having at least 99%, at least 98% at least 95%, at least 90%, at least 85%, at least 80%, at least 75% sequence identity with the VH domain according to SEQ ID NO: 27. In a preferred embodiment the sequence variation does not include the CDR sequences.
In a preferred embodiment according to all aspects of the invention the inhibitor of C5a activity is an antibody or antigen-binding fragment thereof that specifically binds to human C5a comprises (i) a light chain variable domain (VL) according to SEQ ID NO: 22 and (ii) a heavy chain variable domain (VH) according to SEQ ID NO: 27.
In a preferred embodiment according to all aspects of the invention the inhibitor of C5a activity is an antibody or antigen-binding fragment thereof that specifically binds to human C5a comprises (i) a light chain variable domain (VL) having at least 99%, at least 98% at least 95%, at least 90%, at least 85%, at least 80%, at least 75% sequence identity with the VL domain according to SEQ ID NO: 31 and (ii) a heavy chain variable domain (VH) having at least 99%, at least 98% at least 95%, at least 90%, at least 85%, at least 80%, at least 75% sequence identity with the VH domain according to SEQ ID NO: 36. In a preferred embodiment the sequence variation does not include the CDR sequences.
In a preferred embodiment according to all aspects of the invention the inhibitor of C5a activity is an antibody or antigen-binding fragment thereof that specifically binds to human C5a comprises (i) a light chain variable domain (VL) according to SEQ ID NO: 31 and (ii) a heavy chain variable domain (VH) according to SEQ ID NO: 36.
In a preferred embodiment according to all aspects of the invention the inhibitor of C5a activity is an antibody or antigen-binding fragment thereof that specifically binds to human C5a comprises (i) a light chain variable domain (VL) having at least 99%, at least 98% at least 95%, at least 90%, at least 85%, at least 80%, at least 75% sequence identity with the VL domain according to SEQ ID NO: 44 and (ii) a heavy chain variable domain (VH) having at least 99%, at least 98% at least 95%, at least 90%, at least 85%, at least 80%, at least 75% sequence identity with the VH domain according to SEQ ID NO: 52. In a preferred embodiment the sequence variation does not include the CDR sequences.
In a preferred embodiment according to all aspects of the invention the inhibitor of C5a activity is an antibody or antigen-binding fragment thereof that specifically binds to human C5a comprises (i) a light chain variable domain (VL) according to SEQ ID NO: 44 and (ii) a heavy chain variable domain (VH) according to SEQ ID NO: 52.
In a preferred embodiment according to all aspects of the present invention the inhibitors of C5a activity are antibodies or antigen binding fragments thereof disclosed herein comprising one, two or three modified amino acids within the CDR sequences. The modified amino acids are selected from substitution, insertion or deletion, preferably substitution. In a preferred embodiment the one, two or three modified amino acids are conservative amino acid substitutions. “Conservative substitutions” may be made, for instance, on the basis of similarity in polarity, charge, size, solubility, hydrophobicity, hydrophilicity, and/or the amphipathic nature of the amino acid residues involved. Amino acids can be grouped into the following six standard amino acid groups:
(1) hydrophobic: Met, Ala, Vai, Leu, He;
(2) neutral hydrophilic: Cys, Ser, Thr, Asn, Gin;
(3) acidic: Asp, Glu;
(4) basic: His, Lys, Arg;
(5) residues that influence chain orientation: Gly, Pro; and
(6) aromatic: Trp, Tyr, Phe.
As used herein, “conservative amino acid substitutions” are defined as exchanges of an amino acid by another amino acid listed within the same group of the six standard amino acid groups shown above. For example, the exchange of Asp by Glu retains one negative charge in the so modified polypeptide. In addition, glycine and proline may be substituted for one another based on their ability to disrupt a-helices. Some preferred conservative substitutions within the above six groups are exchanges within the following sub-groups: (i) Ala, Vai, Leu and He; (ii) Ser and Thr; (ii) Asn and Gin; (iv) Lys and Arg; and (v) Tyr and Phe. Given the known genetic code, and recombinant and synthetic DNA techniques, the skilled scientist readily can construct DNAs encoding the conservative amino acid variants.
The inhibitors of C5a activity disclosed herein may freely be combined with the inhibitors of IL-6 activity for the claimed use.
In a preferred embodiment the inhibitor of C5a activity is vilobelimab and the inhibitor of IL-6 activity is tocilizumab.
Infectious pneumonia and infectious acute respiratory distress syndrome (ARDS)
Pneumonia is a medical condition of the lung characterized by inflammation of the lung tissue in particular the aleveoli. Pneumonia can be caused by various pathogens, including bacteria, viruses, fungi, and other microorganisms. The infection causes the alveoli in the lungs to become filled with fluid, pus, and cellular debris, which impairs the lungs' ability to efficiently exchange oxygen and carbon dioxide.
In a preferred embodiment the infectious pneumonia is viral pneumonia, bacterial pneumonia or fungal pneumonia, preferably viral pneumonia or bacterial pneumonia, more preferably viral pneumonia. Viral pneumonia may be caused by various viruses including but not limited to influenza virus A or B, respiratory syncytial virus, and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Preferably the infectious viral pneumonia is caused by SARS-CoV-2.
In a more preferred embodiment the infectious pneumonia is COVID-19 pneumonia, i.e. viral pneumonia caused by SARS-CoV-2 infection.
Acute respiratory distress syndrome (ARDS) is a severe and life-threatening condition that affects the lungs. ARDS is characterized by inflammation and damage to the alveoli. This damage results in increased permeability of the alveolar-capillary membrane, causing fluid leakage into the alveoli impairing their ability to effectively exchange oxygen and carbon dioxide.
In a preferred embodiment the ARDS is viral ARDS or bacterial ARDS. In a more preferred embodiment the viral ARDS is COVID-19 ARDS.
Administration regimen
In a preferred embodiment the inhibitor of C5a activity or the inhibitor of IL-6 activity are for administration in a regime comprising an induction phase followed by a maintenance phase. Preferably, the inhibitor of C5a activity is administered in the induction phase and the maintenance phase and the inhibitor of IL-6 activity is administered:
(I) within 0-30 days, preferably within 0-15 days, more preferably within 0-7 days, most preferably within 0-1 days, prior to the induction phase,
(II) during the induction phase, and/or
(III) during the maintenance phase.
In a preferred embodiment, an additional second dose of the inhibitor of IL-6 activity is administered according to (I), (II) or (III).
In a preferred embodiment, the inhibitor of C5a activity is administered in the induction phase and the maintenance phase and the inhibitor of IL-6 activity is administered within 1 to 4 weeks, preferably within 1-2 weeks, more preferably within 1 week, prior to the induction phase.
In a preferred embodiment, the inhibitor of C5a activity is administered in the induction phase and the maintenance phase and the inhibitor of IL-6 activity is administered 1 to 14 days, preferable 1 to 7 days, prior to the induction phase.
In a preferred embodiment, the inhibitor of C5a activity is administered in the induction phase and the maintenance phase and the inhibitor of IL-6 activity is administered during the induction phase.
In a preferred embodiment, the inhibitor of C5a activity is administered in the induction phase and the maintenance phase and the inhibitor of IL-6 activity is administered during the maintenance phase.
In a preferred embodiment, the inhibitor of C5a activity is administered before the inhibitor of IL-6 activity
In a preferred embodiment the induction phase has a duration of 1 to 10 days, preferably 2 to 8 days, more preferably 8 days.
In a preferred embodiment during the induction phase 1 to 8 doses, preferably 2 to 6 doses, more preferably 3 to 5 doses, most preferably 4 doses, of the inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab) are administered.
In a preferred embodiment, each dose administered in the induction phase comprises 200-1600mg, preferably 400-1200mg, more preferably 600-1000mg, most preferably 800mg, of the inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab).
In a preferred embodiment the inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab) is administered by intravenous infusion or injection, preferably infusion.
In a preferred embodiment the maintenance phase has a duration of 1 to 4 weeks, preferably 2-3 weeks, most preferably 2 weeks.
In a preferred embodiment during the maintenance phase, the inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab) is administered every 1 to 4 weeks, preferably 1 to 2 weeks, more preferably every week.
In a preferred embodiment, each dose administered in the maintenance phase comprises 200-1600mg, preferably 400-1200mg, more preferably 600-1000mg, most preferably 800mg, of the inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab).
In a preferred embodiment, the inhibitor of C5a activity and the inhibitor of IL-6 activity are used in an administration regimen, wherein
(i) the induction phase comprises or essentially consists of a period of 1 to 10 days, preferably 2 to 8 days, more preferably 7-8 days, wherein 1-8 doses, preferably 2-4 doses, comprising each 200-1600 mg, preferably 400-1200 mg of said inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab), are administered; and
(ii) the maintenance phase comprises or essentially consists of a period of at least 1 week, preferably 2-4 weeks, wherein a dose, comprising 200-1600 mg, preferably 400-1200 mg of said inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab), is administered every 1-4 weeks, preferably every week.
In a preferred embodiment, the inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab) is administered within the induction and the
maintenance phase at a total maximum of 4 to 12 doses, preferably 6 to 10 doses, more preferably 6 to 8 doses, most preferably 6 doses.
In a preferred embodiment, the inhibitor of C5a activity and the inhibitor of IL-6 activity are used in an administration regimen, wherein
(i) the induction phase essentially consists of a period of 8 days, wherein 4 doses comprising each 800 mg of the inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab), are administered (preferably on days 1, 2, 4 and 8); and
(ii) the maintenance phase essentially consists of a period of 2 weeks, wherein a dose, comprising 800 mg of the inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab), is administered every 1 week (preferably on day 15 and 22 of the treatment).
In a preferred embodiment, the inhibitor of IL-6 activity (preferably tocilizumab) is administered at a dose of 200-1600mg per dose, preferably 400-1200mg, more preferably 600- lOOOmg, most preferably 800mg.
In a preferred embodiment, the inhibitor of IL-6 activity (preferably tocilizumab) is administered at a dose of 2 mg/kg to 12 mg/kg, preferably 3 mg/kg to 10 mg/kg, more preferably 4 mg/kg to 8 mg/kg.
In a preferred embodiment, the inhibitor of IL-6 activity (preferably tocilizumab) is administered at a weight dependent dose of 8mg/kg for patients at or above 30 kg weight or 12 mg/kg for patients of less than 30 kg weight.
In a preferred embodiment, the inhibitor of IL-6 activity (preferably tocilizumab) is administered at a maximum dose of 800 mg per administration.
In a preferred embodiment, the inhibitor of IL-6 activity (preferably tocilizumab) is administered in 1 to 4 doses, preferably 1-2 doses.
In a preferred embodiment, the inhibitor of IL-6 activity (preferably tocilizumab) is administered as a single dose.
In a preferred embodiment, the inhibitor of IL-6 activity (preferably tocilizumab) is administered as a first and second dose. A second dose is in particular beneficial if clinical signs or symptoms do not improve after the first dose. Preferably, the additional dose is at administered at least 8 hours after the first dose.
In a preferred embodiment, each further dose of the inhibitor of IL-6 activity (preferably tocilizumab) is administered at least 8 hours after the previous administration.
In a preferred embodiment, an additional dose of the inhibitor of IL-6 activity (preferably tocilizumab) is administered 8-24 hours after the first dose.
In a preferred embodiment, the inhibitor of IL-6 activity (preferably tocilizumab) is administered every 1 to 4 weeks, preferably every 1 to 2 weeks, preferably weekly.
In a preferred embodiment, the inhibitor of IL-6 activity (preferably tocilizumab) is administered by intravenous infusion or injection, preferably infusion.
In a preferred embodiment, each dose of the inhibitor of IL-6 activity (preferably tocilizumab) is administered prior to the induction phase.
In a preferred embodiment, the inhibitor of IL-6 activity (preferably tocilizumab) is administered as 1-2 doses, preferably single dose, prior (preferably 1-2 weeks prior, more preferably within 1 week prior) to the induction phase.
In a preferred embodiment, the inhibitor of IL-6 activity (preferably tocilizumab) is administered within the induction phase.
In a preferred embodiment, the inhibitor of IL-6 activity (preferably tocilizumab) is administered in more than a single dose, wherein at least one dose is administered prior to the induction phase and at least one dose is administered in the induction phase and/or maintenance phase.
In a preferred embodiment, the inhibitor of IL-6 activity (preferably tocilizumab) is administered in more than a single dose, wherein at least one dose is administered prior to the induction phase and at least one dose is administered in the induction phase.
In a preferred embodiment, the inhibitor of IL-6 activity (preferably tocilizumab) is administered in more than a single dose, wherein at least one dose is administered in the induction phase and at least one dose is administered in the maintenance phase.
In a preferred embodiment, the inhibitor of C5a activity and the inhibitor of IL-6 activity are used in an administration regimen, wherein
(i) 1 or 2 doses (preferably single dose) of the inhibitor of IL-6 activity (preferably tocilizumab) is administered within 0 to 14 days prior and/or after the start of the induction phase (preferably within 0 to 7 days (i.e. prior or after) of the start of the induction phase, more preferably within 24 hours (i.e. prior or after) of the start of the induction phase), preferably the inhibitor of IL-6 activity (preferably tocilizumab) is administered at a weight dependent dose of 8mg/kg for patients at or above 30 kg weight or 12 mg/kg for patients of less than 30 kg weight;
(ii) the induction phase comprises or essentially consists of a period of 1 to 10 days, preferably 2 to 8 days more preferably 7-8 days, wherein 1-8 doses, preferably 2-4 doses, comprising each 200-1600 mg, preferably 400-1200 mg of the inhibitor of C5a activity
(preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab), are administered; and
(iii) the maintenance phase comprises or essentially consists of a period of at least 2 weeks, preferably 2-4 weeks, wherein a dose, comprising 200-1600 mg, preferably 400-1200 mg of said inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab), is administered every 1-4 weeks, preferably every week.
In a preferred embodiment, the inhibitor of C5a activity and the inhibitor of IL-6 activity are used in an administration regimen, wherein
(i) the induction phase comprises or essentially consists of a period of 1 to 10 days, preferably 2 to 5 days, wherein 1-8 doses, preferably 2-4 doses, comprising each 200-1600 mg, preferably 400-1200 mg of the inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab), are administered;
(ii) 1 or 2 doses (preferably single dose) of the inhibitor of IL-6 activity (preferably tocilizumab) is administered within the induction phase; and
(iii) the maintenance phase comprises or essentially consists of a period of at least 2 weeks, preferably 2-4 weeks, wherein a dose, comprising 200-1600 mg, preferably 400-1200 mg of said inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab), is administered every 1-4 weeks, preferably every week.
In a preferred embodiment, the inhibitor of C5a activity and the inhibitor of IL-6 activity are used in an administration regimen, wherein
(i) 1 or 2 doses (preferably single dose) of the inhibitor of IL-6 activity (preferably tocilizumab) is administered 1-2 weeks (preferably within 1 week of start of induction phase) prior to the induction phase;
(ii) the induction phase comprises or essentially consists of a period of 1 to 10 days, preferably 2 to 5 days, wherein 1-8 doses, preferably 2-4 doses, comprising each 200-1600 mg, preferably 400-1200 mg of the inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab), are administered;
(iii) 1 or 2 doses (preferably single dose) of the inhibitor of IL-6 activity (preferably tocilizumab) is administered within the induction phase; and
(iv) the maintenance phase comprises or essentially consists of a period of at least 2 weeks, preferably 2-4 weeks, wherein a dose, comprising 200-1600 mg, preferably 400-1200 mg of said inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab), is administered every 1-4 weeks, preferably every week.
In a preferred embodiment, the inhibitor of C5a activity and the inhibitor of IL-6 activity are used in an administration regimen, wherein
(i) the induction phase comprises or essentially consists of a period of 1 to 10 days, preferably 2 to 5 days, wherein 1-8 doses, preferably 2-4 doses, comprising each 200-1600 mg, preferably 400-1200 mg of the inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab), are administered;
(ii) 1 or 2 doses (preferably single dose) of the inhibitor of IL-6 activity (preferably tocilizumab) is administered within the induction phase;
(iii) the maintenance phase comprises or essentially consists of a period of at least 2 weeks, preferably 2-4 weeks, wherein a dose, comprising 200-1600 mg, preferably 400-1200 mg of said inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab), is administered every 1-4 weeks, preferably every week; and
(iv) 1 or 2 doses (preferably single dose) of the inhibitor of IL-6 activity (preferably tocilizumab) is administered within the maintenance phase.
In a preferred embodiment, the inhibitor of C5a activity and the inhibitor of IL-6 activity are used in an administration regimen, wherein
(i) 1 or 2 doses (preferably single dose) of the inhibitor of IL-6 activity (preferably tocilizumab) is administered within 0 to 14 days prior and/or after the start of the induction phase (preferably within 0 to 7 days (i.e. prior or after) of the start of the induction phase);
(ii) the induction phase comprises or essentially consists of a period of 1 to 10 days, preferably 2 to 5 days, wherein 1-8 doses, preferably 2-4 doses, comprising each 200-1600 mg, preferably 400-1200 mg of the inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab), are administered;
(iii) 1 or 2 doses (preferably single dose) of the inhibitor of IL-6 activity (preferably tocilizumab) is administered within the induction phase;
(iv) the maintenance phase comprises or essentially consists of a period of at least 2 weeks, preferably 2-4 weeks, wherein a dose, comprising 200-1600 mg, preferably 400-1200 mg of said inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab), is administered every 1-4 weeks, preferably every week; and
(v) 1 or 2 doses (preferably single dose) of the inhibitor of IL-6 activity (preferably tocilizumab) is administered within the maintenance phase.
In a preferred embodiment, the inhibitor of C5a activity and the inhibitor of IL-6 activity are used in an administration regimen, wherein
(i) the induction phase comprises or essentially consists of a period of 1 to 10 days, preferably 2 to 5 days, wherein 1-8 doses, preferably 2-4 doses, comprising each 200-1600 mg, preferably 400-1200 mg of the inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab), are administered;
(ii) the maintenance phase comprises or essentially consists of a period of at least 2 weeks, preferably 2-4 weeks, wherein a dose, comprising 200-1600 mg, preferably 400-1200 mg of said inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab), is administered every 1-4 weeks, preferably every week; and
(iii) 1 or 2 doses (preferably single dose) of the inhibitor of IL-6 activity (preferably tocilizumab) is administered within the maintenance phase.
In a preferred embodiment, the inhibitor of C5a activity and the inhibitor of IL-6 activity are used in an administration regimen, wherein
(i) 1 or 2 doses (preferably single dose) of the inhibitor of IL-6 activity (preferably tocilizumab) is administered within 0 to 14 days prior and/or after the start of the induction phase (preferably within 0 to 7 days (i.e. prior or after) of the start of the induction phase);
(ii) the induction phase comprises or essentially consists of a period of 1 to 10 days, preferably 2 to 5 days, wherein 1-8 doses, preferably 2-4 doses, comprising each 200-1600 mg, preferably 400-1200 mg of the inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab), are administered;
(iii) the maintenance phase comprises or essentially consists of a period of at least 2 weeks, preferably 2-4 weeks, wherein a dose, comprising 200-1600 mg, preferably 400-1200 mg of said inhibitor of C5a activity (preferably vilobelimab, INab708 or IFX-2, more preferably vilobelimab), is administered every 1-4 weeks, preferably every week; and
(iv) 1 or 2 doses (preferably single dose) of the inhibitor of IL-6 activity (preferably tocilizumab) is administered within the maintenance phase.
In a preferred embodiment, the inhibitor of C5a activity and the inhibitor of IL-6 activity are used in an administration regimen, wherein
(i) 1 dose of the inhibitor of IL-6 activity (preferably tocilizumab) is administered within 24 hours (i.e. prior or after) of the first administration of the inhibitor of C5a activity (preferably vilobelimab); optionally a second dose of the inhibitor of IL-6 activity (preferably tocilizumab) is administered, preferably after at least 8 hours of the first dose of the inhibitor of IL-6 activity; wherein the inhibitor of IL-6 activity (preferably tocilizumab) is administered at a weight dependent dose of 8mg/kg for patients at or above 30 kg weight or 12 mg/kg for patients of less than 30 kg weight;
(ii) the inhibitor of C5a activity (preferably vilobelimab) is administered on day 1, day 2, day 4, day 8, day 15 and day 22, wherein the inhibitor of C5a activity (preferably vilobelimab) is administered at a dose of 800 mg.
Group of patients
In a preferred embodiment the inhibitor of C5a activity or the inhibitor of IL-6 activity are for use in treating patients that are on invasive mechanical ventilation, preferably not more than 48h post intubation upon first administration of the inhibitor of C5a activity.
In a preferred embodiment the inhibitor of C5a activity or the inhibitor of IL-6 activity are for use in treating patients that have a Path / FiCL ratio of between 50 and 300, preferably between 60 and 200.
The ratio of partial pressure of oxygen in arterial blood (Path) to the fraction of inspiratory oxygen concentration (FiCL) is a clinical indicator of hypoxaemia. PaCL/FiCL ratio is used inter alia to classify severity of acute respiratory distress syndrome (ARDS). In a preferred embodiment the following classification of ARDS severity is used: mild (PaCL/FiCL ratio 200-300), moderate (PaCL/FiCL ratio 100-200) and severe (PaCL/FiCL ratio <100).
In a preferred embodiment the inhibitor of C5a activity or the inhibitor of IL-6 activity are for use in treating patients with mild to severe (preferably moderate to severe) ARDS.
Kit of parts
In a third aspect, the present invention provides a kit of parts comprising an inhibitor of C5a activity (preferably vilobelimab) and an inhibitor of IL-6 activity (preferably tocilizumab). Preferably, the kit is further comprising instructions for the use of the inhibitor of C5a activity (preferably vilobelimab) and the inhibitor of IL-6 activity (preferably tocilizumab) and/or means for the application of the compounds.
In a preferred embodiment the kit of parts comprises the inhibitor of C5a activity (preferably vilobelimab) in a dosage form comprising 100-2000 mg, 200-1500mg, 400- 1200mg, preferably 600-1000 mg, more preferably 800mg of an inhibitor of C5a activity (preferably vilobelimab). Preferably wherein the dosage form is a lyophilized powder, or a liquid dosage form, preferably a solution for infusion or a solution for injection.
In a preferred embodiment the kit of parts comprises the inhibitor of IL-6 activity (preferably tocilizumab) in a dosage form comprising 100-2000 mg, 200-15 OOmg, 400- 1200mg, preferably 600-1000 mg, more preferably 800mg of an inhibitor of IL-6 activity
(preferably tocilizumab). Preferably wherein the dosage form is a lyophilized powder, or a liquid dosage form, preferably a solution for infusion or a solution for injection.
Method of treatment
In a further aspect of the invention a method for the treatment of patients with an infectious pneumonia and/or infectious acute respiratory distress syndrome (ARDS), wherein the method is comprising administering to the patient an effective amount of an inhibitor of C5a activity and an inhibitor of IL-6 activity. In a preferred embodiment the method comprises administering the inhibitor of IL-6 activity prior and/or concomitant with the inhibitor of C5a- activity.
In a third aspect, the present invention provides the use of an inhibitor of C5a activity and an inhibitor of IL-6 activity for the manufacturing of a medicament for the treatment of infectious pneumonia and/or infectious acute respiratory distress syndrome (ARDS).
Examples
The following Examples are provided for further illustration of the invention. The invention, however, is not limited thereto, and the following Examples merely show the practicability of the invention on the basis of the above description.
Example 1: Phase II/III study evaluating IFX-1 for the treatment of COVID 19 related severe pneumonia
A) Rationale for the study
Neutrophil driven tissue and organ damage is known to play an important role in a wide array of acute inflammatory diseases. The mechanism leading to damage has been largely attributed to two mechanisms: 1) the release of granular enzymes and 2) the generation of so- called reactive oxygen species in which O2 radical formation elicits a damaging effect.
One of the strongest chemoattractant substances, which is also capable of inducing both mechanisms described above is the human complement split product C5a.
Earlier research by applicant has demonstrated that the anti-human C5a antibody IFX- 1 could significantly reduce the neutrophil and macrophage infiltration in a model of viral (H7N9)-induced lung injury in monkeys (Sun et al. 2015). This was accompanied by a significantly reduced tissue damage in the lung of infected animals and also a significantly reduced viral replication when compared to mock-treated animals. The mechanism was then
confirmed in a model of MERS-CoV-induced lung injury where similar findings were made by using an anti-C5a receptor antibody (Jiang et al. 2018) and in a model of SARS-CoV- induced lung injury by using complement C3 knockout mice (Gralinski et al. 2018). The latter study also provided evidence for a profound complement activation in SARS virus infections in animals. These studies all suggested a role of neutrophil driven lung damage, which was induced by generation of C5a in different viral-lung injury models, including SARS viruses.
Latest data from a collaborating research group in China demonstrated strongly and significantly elevated C5a levels in severely diseased COVID-19 patients when compared to mildly diseased patients (Gao et al. 2020; Carvelli et al. 2020) and revealed evidence for a strong complement pathway driven activation of C5a in this disease. In addition, data from the first two severely diseased COVID 19 patients treated with the IFX-1 cell line derived anti-C5a antibody BDB-1, which InflaRx licensed to a collaborator in China, demonstrated clinical improvement in oxygenation index, fever reduction, and laboratory parameter normalization including liver enzymes and lymphocyte counts.
From a clinical perspective, various papers have confirmed that COVID-19 infected non-surviving patients demonstrated lung failure with close to 100% (Zhou et al. 2020) and that, in contrast to surviving patients, non-survivors demonstrated elevated white blood cell counts and neutrophils above the normal range (Wang et al. 2020; Gong et al. 2020).
In summary, there is evidence that in COVID-19 patients who are severely affected, C5a activation occurs to a large extent and that neutrophil count elevation in blood is associated with bad outcome. These human data fit well to the scientific rationale developed in animal models of viral-induced lung injury that activated neutrophils attracted to the lung may cause viral-induced lung damage. First treatment attempts with the anti-C5a antibody technology developed by InflaRx (BDB-1) in severely affected COVID-19 patients provide further evidence for the scientific rationale.
The completed Phase II portion of the current trial was exploratory in nature and was not powered to show statistically significant differences in clinical endpoints. Relative change (%) from baseline to day 5 in the oxygenation index, chosen as the primary endpoint for Phase II, showed a large variability and dependency on patient positioning and intubation status which excluded this endpoint from being used in a confirmatory study. Phase III of the study is an adequately powered, placebo-controlled, double blinded phase evaluating standard of care (SOC) + IFX-1 versus SOC + placebo-to-match with 28-day all-cause mortality as the primary endpoint, an accepted regulatory primary endpoint for critical care studies.
At the time that Phase II of this study was initiated, there was no SOC established for this newly identified disease. The SOC to be utilized in Phase III of this study reflects the current understanding of the SOC for hospitalized patients with COVID-19, which includes venous thromboembolism (VTE) prophylaxis at a minimum, and may include other international and country-specific recommended treatments for COVID-19 per the locally adopted treatment recommendations.
Rationale for dosage regimen
The aim for determining the dose and administration frequency for IFX-1 is to establish a pragmatic administration schedule for the COVID-19 patient population. IFX-1 dose and administration schedule was chosen based on prior pharmacokinetic (PK)/pharmacodynamic (PD) observations for IFX-1 trough values and blood C5a levels, dose-response assessment in Study IFX-1 in Hidradentis suppurativa, and the described unprecedented high C5a levels in patients with severe COVID-19 related pneumonia (Gao et al. 2020; Carvelli et al. 2020). In a population PK/PD modelling study for IFX 1 based on the data of the previous trials with IFX- 1, a reduction in C5a concentration was found to be dependent on all measures of drug exposure, with increasing exposure predicting a greater reduction in C5a.
Based on the above data, the dosing schedule of IFX-1 for this study includes an additional dose of 800 mg IFX 1 at day 2 in the established fractionated loading dose scheme, that foresees administration of 800 mg at days 1, 4, and 8. This additional dose has been chosen due to the reported high C5a levels in COVID-19 patients (Gao et al. 2020). Therefore, the entire dosing scheme contains up to 6 IFX-1 doses of 800 mg at days 1, 2, 4, 8, 15, and 22 (or less until hospital discharge). This regimen had been employed in Phase II of this study, leading to a lower death rate and supporting efficacy signals without any new signals of unknown toxicities (Vlaar et al 2020).
B) Objectives of the study
Primary Objectives
The primary objective of Phase III is:
• To demonstrate the efficacy of IFX-1 to improve survival outcomes of severe CO VID 19 pneumonia (confirmative)
Secondary Objectives
The secondary objectives of Phase III are:
To assess and define other parameters of efficacy
To assess the safety of IFX-1
C) Study design
Overall design
This is a pragmatic, adaptive, randomized, multicenter phase II/III study evaluating IFX-1 for the treatment of CO VID 19 related severe pneumonia. Phase III is a double-blind, placebo-controlled, randomized phase evaluating SOC + IFX-1 (Arm A) and SOC + placebo- to-match (Arm B). The SOC includes VTE prophylaxis at a minimum, and may include other international and country-specific recommended treatments for COVID-19 per the locally adopted treatment recommendations.
Phase III of the study will randomize up to 400 patients with one interim analysis for stopping for futility. A total of 180 patients are planned to be randomized into Arm A and Arm B using a 1 : 1 allocation ratio for the first stage; based on results of the interim analysis, up to an additional 180 patients are planned to be randomized using the same allocation ratio for the second stage. Additional patients will be randomized under the following conditions:
• For patients who were erroneously randomized (screening failures) and did not get IMP treatment.
• For patients who were randomized but consent was withdrawn (either by the patient or patient representative) within the first 48 hours after randomization, irrespective of whether IMP was administered or not.
The primary statistical analysis will be based on all randomized patients except patients randomized in error who did not get IMP treatment.
In Phase III, patients will be treated with a maximum of 6 IV doses of IFX-1 800 mg (Arm A) or placebo (Arm B) at days 1, 2, 4, 8, 15, and 22, as long as the patient is still in the hospital (even if discharged from the ICU).
Patients will be assessed for quality of life using the EQ-5D.
Patients will be followed for survival and their clinical status assessed by the Glasgow Outcome Scale.
End of Treatment and Follow-up
End of treatment (EOT) is defined as the last planned administration or discontinuation of IMP for other reasons.
The FUV is scheduled to occur 28 days (if patient has been discharged) and 60 days after the randomization date. The visit information obtained at a site visit or via telephone call (e.g., AEs, survival status, according to the SoA) should be recorded.
Discontinuation of study treatment does not represent withdrawal from the study.
End of Study
The end of study for the individual patient is defined as the date of the last contact of the follow up period (i.e., 60 days after randomization), date of death, date of consent withdrawal from study participation, or the date of last contact when patients are lost to followup, whatever occurs earliest.
The entire study will end when all patients have discontinued IFX 1 AND all patients ended the study as described above.
D) Study population
Inclusion criteria
Patients must meet all the following criteria at randomization to be enrolled into Phase III of the study:
1. At least 18 years of age or older
2. Patient on invasive mechanical ventilation (but not more than 48h post intubation at time point of first IMP administration)
3. Patients with a PaCh / FiCh ratio of < 200 and > 60 at randomization (one representative measurement within 6h before randomization)
4. SARS-CoV-2 infection confirmation (tested positive in last 14 days before randomization with locally available test system)
Exclusion criteria
Patients who fulfill any of the following criteria at randomization are not eligible to participate in Phase III of the study:
1. Intubated > 48 h at time point of first IMP administration
2. Expected stop of invasive ventilation or expected extubation in the next 24h without additional intervention according to judgment of the investigator
3. Known history of chronic dialysis OR received renal replacement therapy in past 14 days OR anticipated to receive renal replacement therapy within 24h after randomization
4. Known history of progressed COPD as evidenced by use of daily maintenance treatment with long-acting bronchodilators or inhaled/oral corticosteroids for > 2 months
5. Treatment of COVID-19 with investigational antibody treatment(s) which are not approved or not included in locally adopted treatment guidelines (e.g., WHO guidance, National Institutes of Health [NIH] CO VID-19 treatment guidelines) for this indication in the past 7 days (Note: Antibody treatments] given within past 7 days for pre existing diseases, other than COVID-19, are allowed.)
6. At time point of randomization, treatment of COVID 19 with investigational treatments which are not approved or not included in locally adopted treatment guidelines for this indication (e.g., WHO guidance, NIH COVID-19 treatment guidelines), including SARS CoV-2 multiplication inhibitor(s) or immunomodulator(s). (Note: If a locally adopted treatment guideline recommends drugs such as remdesivir, dexamethasone, or anticoagulation, this would be allowed. Adopted guidelines and updates must be documented at study initiation and throughout the conduct of the study.)
7. Received cytokine adsorption therapy in past 3 days
8. Known hypersensitivity to IFX-1 or any other ingredient of the study medication
9. Serum or urine pregnancy test positive before randomization (required for women of childbearing potential)
10. Received organ or bone marrow transplantation in past 3 months
11. Known cardio-pulmonary mechanical resuscitation in past 14 days
12. Patient moribund or expected to die in next 24h according to the judgment of the investigator
13. Known to have received anti-cancer therapy for hemato-oncological disease in past 4 weeks OR known to have active malignant disease at time point of randomization
14. Known severe congestive heart failure (corresponding to e.g. NYHA Class III- IV, left ventricular ejection fraction <40%; see Appendix 8)
15. Known history of chronic liver disease (Child-Pugh B or C; see Appendix 11)
16. Participating in or has participated in other investigational interventional studies (drug or device) within the last 7 days before randomization
Screen failures
Screening failures are defined as patients who do not meet the criteria for participation in this study and thus, are either not randomized/assigned to IMP or randomized in error and do/did not receive IMP. Screen failure data for not randomized patients will not be recorded in the eCRF, for those patients who are randomized in error, screen failure data are to be recorded in the eCRF.
Rescreening does not apply to this study.
E) Study intervention
Investigational medicinal product (IMP)
The IMP is defined as the investigational treatment IFX-1 and Placebo-to-match (Placebo) that is intended to be administered to a study patient according to the study protocol.
IFX-1 concentrate solution for infusion will be supplied in 20 mL glass vials at a concentration of 10 mg/mL (200 mg per vial) for reconstitution and IV administration. Apart from IFX 1, the solution will contain sodium chloride, sodium phosphate, and polysorbate 80.
Placebo concentrate solution for infusion will be supplied in 20 mL glass vials for reconstitution and IV administration. The solution will contain sodium chloride, sodium phosphate, and polysorbate 80.
Administration and Dosage
For Phase III, patients are randomized to either receive SOC + IFX-1 in treatment Arm A or SOC + Placebo in treatment Arm B. The first dose of IFX-1 or Placebo is administered at Day 1. SOC may start at any time according to their administration schedule.
Close observation of IFX-1 or Placebo infusion(s) is required for monitoring of potential infusion reactions. Appropriate treatment for potential infusion reactions must be available during this time.
Patients will be treated with a maximum of 6 IV doses of IFX-1 800 mg (Arm A) or placebo (Arm B) administered as a 30-60-minute IV infusion. The 6 treatments are at days 1, 2, 4, 8, 15, and 22 as long as the patient is still in the hospital, even if discharged from ICU. IMP administrations for Phase III are detailed in Figure 1.
Prior and Concomitant Medication/Treatments
In Phase III of the study, in addition to the IMP, all patients will receive SOC for treatment of COVID-19, which includes VTE prophylaxis with anticoagulants at a minimum. Other international or country-specific recommended treatments for COVID-19 per the locally
adopted treatment recommendations (including but not limited to corticosteroids, remdesivir, and other local SOC) are allowed as concomitant medications. SOC treatment start and stop is not defined by the protocol and can start at any time. SOC treatment will be given according to investigator’s discretion.
Reportable medications or vaccinations that the patient is receiving within 7 days prior to randomization and up to 30 days after the last dose of study treatment should be recorded along with:
• Reason for use
• Dates of administration including start and end dates
F) Study assessments and procedures
Baseline Assessments (Screening)
The following procedures will be performed to assess baseline characteristics during screening evaluations and before randomization and IMP administration:
• Check inclusion and exclusion criteria
• Documentation of demographic details (age, gender, race, and ethnicity), medical and COVID- 19 history. Race information is required for eGFR calculation using the CKD-EPI equation
• 8-point ordinal scale assessment and oxygenation index (PaO? / FiO?; one representative measurement within 6h before randomization)
• Documentation of concomitant disease and prior medications
• Baseline safety assessment (height, weight, physical exam, AEs, ECG, urine analysis, pregnancy test, and clinical safety laboratory [including creatinine assessment]))
Prior and Concomitant Medications and Procedures
All procedures performed or medications administered within 7 days before randomization should be recorded as prior procedures and prior medication with generic name, start date, stop date, and indication for treatment.
Reporting of prior and concomitant medications will follow special considerations for conditions in the ICU. A complete record of all prior and concomitant medications (but excluding nutritional and volume therapy, electrolyte support, vitamins, non-steroidal antiinflammatory drugs (NSAIDs), and supportive therapies such as artificial tears, ointments, stool
softeners/laxatives, etc.) will be maintained in the eCRF for each participant, beginning 7 days before randomization and continuing up to 30 days after the last dose of study treatment).
In addition, a complete record of all steroid and antibiotic therapy, as well as any therapy (medications, specific treatments, etc.) associated with or used in the assessment or treatment of an AE will be documented for the duration of the study.
The following information must be recorded in the eCRF for each reportable concomitant medication: generic name, route of administration, start date, stop date, and indication
G) Adverse events
Definition of adverse events (AE):
An AE is any untoward medical occurrence in a subject administered an IMP; an AE does not necessarily have to have a causal relationship with this treatment.
AEs encompass any unfavorable and unintended sign (including an abnormal laboratory finding), symptom, or disease that arises or worsens after the inclusion of the subject into the study.
H) Statistical considerations
Determination of sample size
A total of 180 patients (90 per arm) are planned to be randomized in Stage 1 and up to 180 patients (90 per arm) in Stage 2. For patients randomized in error (screening failures) and patients for whom consent is withdrawn within 48 hours after randomization, additional patients will be randomized. This results in 90% overall power to show efficacy in the final efficacy analysis. The power calculation is based on an overall 2.5% one-sided alpha and an assumed 30% 28-day mortality under Placebo and 15% 28-day mortality under IFX-1 treatment.
Analysis sets
Full analysis set (FAS): The primary statistical analysis will be based on all randomized patients except patients randomized in error (reason for early termination documented as “Randomized by mistake” in the eCRF) who did not get IMP treatment.
Safety analysis set (SAF): Safety analyses will be based on all patients who received at least one infusion of IMP, and patients will be analyzed according to the treatment they actually received.
Statistical Analyses
Primary Efficacy Analysis
The primary analysis will be performed based on the FAS.
The primary efficacy variable is 28-day mortality (proportion of patients deceased until day 28). The primary statistical hypotheses to be tested are:
Ho: HR = 1 versus
Hi: HR > 1 where HR is the hazard ratio derived from the beta coefficient of the primary statistical analysis Cox regression model comparing the SOC + Placebo treatment arm (Arm B) with the SOC + IFX-1 treatment arm (Arm A).
A total of 180 patients are planned to be randomized to Arm A and Arm B using a 1 : 1 allocation ratio for the first stage. The interim analysis is performed after 180 patients, not counting patients being randomized in error, have been followed-up until day 28 (or died before). In case the interim analysis does not result in an early stop for futility, 180 additional patients are planned to be randomized in a ratio of 1 : 1 to Arm A and B. The planned maximum number of patients in the study is 360. Depending on the number of patients meeting criteria for additional randomization, more patients will be randomized but not more than 400.
The primary statistical analysis of IFX-1 + SOC compared to SOC alone will make use of a one-sided alpha level of 2.5% and will test for superiority (lower mortality among IFX-1 treated patients). The primary statistical analysis will be based on a Cox proportional hazards regression model with outcome 28-day all-cause mortality as a censored time-to-event variable and explanatory variables treatment arm (Arm B versus Arm A) and age. The primary statistical analysis will be based on all randomized patients except patients randomized in error (reason for early termination documented as “Randomized by mistake” in the eCRF) who did not get IMP treatment. For the primary analysis, all-cause mortality will be censored at day 28 for subjects who died after day 28 or who have more than 28 days follow-up alive.
The z-statistic for the interim and final analysis will be calculated as the proportion of the beta coefficient for the treatment arm effect and its standard error from the Cox proportional hazards regression model. The study will be stopped for futility if the z-statistic for the first stage is 0 or lower.
Secondary Efficacy Analysis
The secondary efficacy analyses will be performed based on the FAS.
The first secondary efficacy endpoint will only be addressed with statistical hypothesis tests if the primary endpoint is statistically significant using the full overall 2.5% one-sided alpha. If the first secondary endpoint is also statistically significant, the full 2.5% one-sided alpha will be passed to the 4 remaining secondary endpoints. Multiplicity in the secondary endpoints will be addressed with the fallback method. 60-day mortality will be analyzed similar to 28-day mortality and the other secondary endpoints will be evaluated using a logistic regression model with the explanatory variables randomized treatment arm (Arm B versus Arm A) and age for:
1. 60-day all-cause mortality
2. Proportion of patients with an improvement in the provided 8-point ordinal scale at day 15 (at least one score point lower than at randomization)
3. Proportion of patients with an improvement in the provided 8-point ordinal scale at day 28 (at least one score point lower than at randomization)
4. Proportion of patients developing acute kidney failure (eGFR < 15 mL/min/1.73m2) during ICU stay and at day 28
5. Proportion of patients free of any renal replacement therapy within 28 days upon randomization
The ordering of the secondary endpoints 2 to 5 for hypothesis testing will be as in above mentioned list. The full 2.5% one-sided alpha for the fallback method will be attributed to the 4 secondary endpoints in the following way 2%, 0.2%, 0.2%, and 0.1%. If the preceding hypothesis test is not significant, subsequent tests will be performed at the aforementioned alpha level. If tests are significant, the alpha is added to the subsequent hypothesis test (e.g., if the primary hypothesis test is significant and secondary endpoints 1-4 are all significant, the fifth secondary endpoint will be tested at an alpha of 2.5%; if the fourth secondary endpoint is not significant, the fifth secondary endpoint will be tested at an alpha of 0.1%).
The primary endpoint as well as all secondary endpoints will also be evaluated as censored time to event variables by Kaplan-Meier type methods. Kaplan-Meier analyses will be performed comparing the two treatment arms overall and within the following stratifications:
• Stratification by site
• Stratification by country
• Stratification by sex
• Stratification by age
Stratification by comorbidities
Stratification by standard or care.
Other Efficacy Analyses
Time to First Extubation
Time to first extubation will be analyzed as a censored time to event variable. Withdrawing early from the study will lead to the patient being right-censored at the date of withdrawal provided that the reason is not an outcome of interest (extubation or death). In order to adequately account for competing outcomes as well as potential right-censoring due to varying follow-up times, the outcome will be non-parametrically analyzed using Aalen- Johansen-type estimation based on a competing risks model with two absorbing states (event 1 : all-cause death; event 2: extubation). Adjustment for age will be realized by regression modeling accounting for competing risks.
Patients Alive and Free of Respiratory Failure
Patients achieving 8-point ordinal scale score 3 or below (patients alive and free of respiratory failure) at day 15 and day 28 are defined as alive and free of respiratory failure at the respective study day. The corresponding endpoint will be analyzed using logistic regression with explanatory variables age and treatment group.
Glasgow Outcome Scale
Glasgow Outcome Scale assessed at study day 60 will be analyzed via ordinal logistic regression with explanatory variables treatment arm (Arm B versus Arm A) and age.
Quality of Life
Quality of life will be assessed by EQ-5D at study day 60. The visual analogue scale as well as an index value based on the 5 health states (cross-walk index value using the United States value set) will be analyzed by an ANCOVA model with explanatory variables treatment arm (Arm B versus Arm A), age, and sex (male versus female).
Interim Analysis
The interim analysis is performed after the first 180 patients, not counting patients being randomized in error, in Stage 1 have been followed-up until day 28 (or died before). The study will be stopped for futility if the z-statistic of the primary efficacy analysis is 0 or lower. If the
study is not stopped for futility, another up to 180 patients will be randomized in Stage 2. Depending on the number of patients meeting criteria for additional randomization, more than 360 patients will be randomized but not more than a total of 400.
Example 2: Patients receiving combined treatment of IFX-1 and Tocilizumab in Phase III study evaluating IFX-1 for the treatment of COVID 19 related severe pneumonia
A) Prior or concomitant use of Tocilizumab
Some patients enrolled in the phase III trial outlined in example 1 above where also receiving the anti-IL-6 receptor antibody Tocilizumab, which had received an emergency use authorization by the FDA and EMA for the treatment of severe COVID-19.
Unexpectedly, patients receiving both medications, i.e. IFX-1 (vilobelimab) and tocilizumab, exhibited a highly significant reduction of all-cause mortality as compared to placebo treatment as well as single use of vilobelimab or tocilizumab.
This significant reduction was observed in patients receiving tocilizumab prior to first administration of vilobelimab (i.e. tocilizumab baseline use) as well as concomitant use of both compounds.
For the purpose of the present application the following definitions of baseline use and concomitant use will be used
Baseline use of tocilizumab is defined as receiving at least one dose of tocilizumab between study day -7 and study day 1.
Prior use of tocilizumab is defined as receiving at least one dose of tocilizumab before day-7 (i.e. prior to baseline use) concomitant use: Medications will be counted as concomitant either
• if they have an end date/time later than the timestamp of randomization
• or if “ongoing' is ticked on the respective eCRF page.
If the end date/time is completely missing and “ongoing” is not ticked, the previous medication will also be assumed to be concomitant. Incomplete start/end dates will not be imputed. If the incomplete date does not allow clear categorization as previous or concomitant medication, the medication will be counted as concomitant.
For the Cox proportional hazard models and Kaplan-Meier curves reported herein tocilizumab use is either classified as ‘baseline usage of Tocilizumab’ or ‘any usage of Tocilizumab’, whereas the later encompasses prior use, baseline use and concomitant use.
B) Effect of combined treatment with an inhibitor of C5a activity and an inhibitor of IL-6 activity
The primary statistical analysis is based on a Cox proportional hazards regression model with outcome 28-day and 60-day all-cause mortality as a censored time-to-event variable and explanatory variables treatment arm (Arm B (i.e. placebo + SOC) versus Arm A (i.e. vilobelimab + SOC) and age. This statistical analysis was made with patients receiving Tocilizumab (baseline use or any use) in Arm A and or Arm B.
Of note, a hazard ratio (HR) above 1 indicates a covariate that is positively associated with the event probability, and thus negatively associated with the length of survival. In summary, HR = 1 : No effect, HR < 1 : Reduction in the hazard and HR > 1 : Increase in Hazard. Therefore a covariate with a hazard ratio > 1 is a bad prognostic factor and a covariate with a hazard ratio < 1 is called good prognostic factor.
Combined treatment vilobelimab and tocilizumab (any usage)
As shown in Table 2 below, the 28-day all-cause-mortality for patients receiving vilobelimab and tocilizumab (any usage) is significantly reduced as compared to the placebo group (i.e. tocilizumab any usage) (p=0.0098, HR: 0.140). In other words, the combined treatment of vilobelimab and tocilizumab (any usage) is significantly more effective in reducing all cause mortality as compared to tocilizumab alone. All treatment group received in addition to the indicated treatment also standard of care (SOC).
Table 2: Cox proportional hazard model for 28-day all-cause mortality for patients with any usage of Tocilizumab - FAS; (number of patients in analysis set/model: 61/61)
* Wald-type confidence intervals; HR = Hazard Ratio; FAS = Full analysis set; SOC = Standard of care; Vilo = Vilobelimab
The same significant reduction is observed for 60-day all-cause mortality as shown in Table 3 below. The treatment group receiving vilobelimab and tocilizumab (any usage) had a significantly reduced all-cause mortality as compared with the placebo group (i.e. tocilizumab any usage) (p=0.0187; HR: 0.296).
Table 3: Cox proportional hazard model for 60-day all-cause mortality for patients with any usage of Tocilizumab - FAS; (number of patients in analysis set/model: 61/61)
* Wald-type confidence intervals; HR = Hazard Ratio; FAS = Full analysis set; SOC = Standard of care; Vilo = Vilobelimab
Combined treatment vilobelimab and tocilizumab (baseline usage)
A similar effect was observed for baseline usage of Tocilizumab (defined as receiving at least one dose of tocilizumab between study day -7 and study day 1).
The 28-day all-cause mortality was dramatically reduced if the patients received baseline usage of Tocilizumab in combination with vilobelimab as indicated by the hazard ratio of 0.0.073 (p=0.012; see Table 4 below).
Table 4: Cox proportional hazard model for 28-day all-cause mortality for patients with baseline usage of Tocilizumab - FAS; (number of patients in analysis set/model: 60/60);
* Wald-type confidence intervals; HR = Hazard Ratio; FAS = Full analysis set; SOC = Standard of care; Vilo = Vilobelimab
A similar effect was observed for 60-day all-cause mortality after Tocilizumab baseline usage. The all-cause mortality was dramatically reduced as indicated by the hazard ratio of 0.245 (p=0.0127; see Table 5 below).
Table 5: Cox proportional hazard model for 60-day all-cause mortality for patients with baseline usage of Tocilizumab - FAS; (number of patients in analysis set/model: 60/60) * Wald-type confidence intervals; HR = Hazard Ratio; FAS = Full analysis set; SOC = Standard of care; Vilo = Vilobelimab
Treatment with vilobelimab without any usage of tocilizumab
Patients treated with vilobelimab alone also had a reduced 28-day all-cause mortality as compared to placebo (no tocilizumab use). However, the effect, as indicated by the hazard ratio of 0.793, was much smaller than the combination of vilobelimab and tocilizumab (HR=0.140).
Table 6: Cox proportional hazard model for 28-day all-cause mortality for patients without any usage of Tocilizumab - FAS; (number of patients in analysis set/model: 307/307) * Wald-type confidence intervals; HR = Hazard Ratio; FAS = Full analysis set; SOC = Standard of care; Vilo = Vilobelimab
The same was observed for 60-day all-cause mortality. Here all-cause mortality was also reduced, but with a similar hazard ratio of 0.756 (see Table 7 below) as compared to the combination of vilobelimab and tocilizumab (HR=0.296).
Table 7: Cox proportional hazard model for 60-day all-cause mortality for patients without any usage of Tocilizumab - FAS; (number of patients in analysis set/model: 61/61) * Wald-type confidence intervals; HR = Hazard Ratio; FAS = Full analysis set; SOC = Standard of care; Vilo = Vilobelimab
Essentially the same result was achieved in the treatment group not receiving baseline usage of Tocilizumab. For the sake of clarity, these group of patients (i.e. no baseline use of Tocilizumab) did not receive any Tocilizumab administration (neither baseline nor any other use), but rather vilobelimab or placebo treatment. The all-cause mortality for 28-days and 60- days was reduced by vilobelimab treatment as indicated by the hazard ratios of 0.801 and 0.763 respectively (see Tables 8 and 9 below).
Table 8: Cox proportional hazard model for 28-day all-cause mortality for patients without baseline usage of Tocilizumab - FAS; (number of patients in analysis set/model: 308/308)
* Wald-type confidence intervals; HR = Hazard Ratio; FAS = Full analysis set; SOC = Standard of care; Vilo = Vilobelimab
Table 9: Cox proportional hazard model for 60-day all-cause mortality for patients without baseline usage of Tocilizumab - FAS; (number of patients in analysis set/model: 308/308)
* Wald-type confidence intervals; HR = Hazard Ratio; FAS = Full analysis set; SOC = Standard of care; Vilo = Vilobelimab
Baseline usage of tocilizumab and interaction with vilobelimab treatment
Further Cox proportional hazard models as a censored time-to-event variable and explanatory variables treatment arm and age and baseline tocilizumab use flag and its interaction with study treatment were established.
In a first analysis the 28-day all-cause mortality with baseline tocilizumab was investigated. As indicated in table 10 below, treatment with vilobelimab alone (i.e. “Treatment (Vilo vs Placebo) for baseline tocilizumab use: no") had only a small effect on 28 day survival rate with a HR of 0.797. Likewise the baseline use of tocilizumab without vilobelimab ('Baseline Tocilizumab use (yes vs no) for Treatment: Placebo") had a minor effect on survival rate with a HR of 0.844.
In contrast the interaction of vilobelimab treatment with baseline use of tocilizumab ^Treatment (Vilo vs Placebo) for baseline tocilizumab use: yes”) resulted in a robust effect on survival with a HR of only 0.073. Consistently, the same robust effect on survival was observed for baseline Tocilizumab use interaction with vilobelimab treatment with a HR of 0.077. Taken together the combined treatment of vilobelimab and baseline tocilizumab resulted in a dramatic increase of survival whereas the treatment with vilobelimab or tocilizumab alone only had a minor effect.
Table 10: Cox proportional hazards regression model with outcome 28-day all-cause mortality as a censored time-to-event variable and explanatory variables treatment arm and age and baseline tocilizumab use flag and its interaction with study treatment - FAS. (number of patients in analysis set/model: 368/368)
* Wald-type confidence intervals; HR = Hazard Ratio; FAS = Full analysis set; SOC = Standard of care; Vilo = Vilobelimab
As similar effect of the respective treatments was observed for 60-day all-cause mortality as summarized in Table 11 below.
Table 11: Cox proportional hazards regression model with outcome 60-day all-cause mortality as a censored time-to-event variable and explanatory variables treatment arm and age and baseline tocilizumab use flag and its interaction with study treatment - FAS. (number of patients in analysis set/model: 368/368)
* Wald-type confidence intervals; HR = Hazard Ratio; FAS = Full analysis set; SOC = Standard of care; Vilo = Vilobelimab
Any usage of tocilizumab and interaction with vilobelimab treatment
The statistical analysis including interaction of the different treatments for any usage of Tocilizumab on 28-day all-cause mortality had a similar result as disclosed above for baseline usage of Tocilizumab (see Table 12 below). The treatment with only vilobelimab ‘Treatment (Vilo vs Placebo) for any tocilizumab use: no had in this group of patient"") had in this group of patients a minor effect as indicated by a HR of 0.789. Likewise treatment with only tocilizumab (any usage) ( ny Tocilizumab use (yes vs no) for Treatment: Placebo’"') had a minor effect as indicated by a HR of 0.844.
In contrast and consistent with the observation for baseline usage of Tocilizumab the interaction of vilobelimab treatment with baseline use of tocilizumab (‘Treatment (Vilo vs Placebo) for any tocilizumab use: yes’" resulted in a robust effect on survival with a HR of only
0.249. Consistently, the same robust effect on survival was observed for any Tocilizumab use interaction with vilobelimab treatment with a HR of 0.276.
Table 12: Cox proportional hazards regression model with outcome 28-day all-cause mortality as a censored time-to-event variable and explanatory variables treatment arm and age and any tocilizumab use flag and its interaction with study treatment - FAS. (number of patients in analysis set/model: 368/368)
* Wald-type confidence intervals; HR = Hazard Ratio; FAS = Full analysis set; SOC = Standard of care; Vilo = Vilobelimab
A virtually identical result was observed for 60-day all-cause mortality as disclosed in Table 13 below.
Table 13: Cox proportional hazards regression model with outcome 60-day all-cause mortality as a censored time-to-event variable and explanatory variables treatment arm and age and any tocilizumab use flag and its interaction with study treatment - FAS. (number of patients in analysis set/model: 368/368)
* Wald-type confidence intervals; HR = Hazard Ratio; FAS = Full analysis set; SOC = Standard of care; Vilo = Vilobelimab
C) Effect of combined treatment of vilobelimab and tocilizumab on patient survival
In addition to the statistical analysis using Cox proportional hazard model the single and combined treatment of vilobelimab and tocilizumab was also analyzed with the Kaplan-Meier method. This is a non-parametric method used to estimate the survival probability from observed survival times and is typically plotted as Kaplan-Meier survival graphs.
The effect size of combined treatment of vilobelimab and tocilizumab (any usage) on all-cause mortality is dramatically increased as can be seen in figure 2. Treatment of vilobelimab combined with any usage of tocilizumab (line 1) shows only a slight increase of the probability of mortality within 60 days. In comparison, the group receiving placebo without any use of Tocilizumab (line 4) had a much higher mortality rate. The mortality rate in the groups receiving vilobelimab and standard of care (SOC) without tocilizumab (line 2) showed
only a slight improvement. Patients treated with tocilizumab and SOC (line 3) showed an even lower improvement of survival rates. A virtually identical outcome was observed with baseline use of Tocilizumab (see figure 3).
The benefit of combined vilobelimab and tocilizumab treatment is even more pronounced within the first 22 days of treatment with hardly any fatalities in this treatment group. In contrast in the other treatment groups the majority of fatalities occurred in this time of treatment.
In summary, the combined treatment of vilobelimab and tocilizumab (baseline use and any use) resulted in a dramatic increase of survival probability as compared to the treatment with only one of the compounds or placebo. Of note the effect on survival rate of the combined treatment is much higher than the sum of the individual effects of vilobelimab and tocilizumab alone, indicating a synergistic effect of the combined treatment instead of a mere additive effect.
Claims
1. An inhibitor of C5a activity for use in:
(i) combination with an inhibitor of IL-6 activity in the treatment of infectious pneumonia and/or infectious acute respiratory distress syndrome (ARDS); and/or
(ii) the treatment of infectious pneumonia and/or infectious acute respiratory distress syndrome (ARDS), wherein the patient is or has been treated with an inhibitor of IL-6 activity.
2. An inhibitor of IL-6 activity for use in:
(i) combination with an inhibitor of C5a activity in the treatment of infectious pneumonia and/or infectious acute respiratory distress syndrome (ARDS); and/or
(ii) the treatment of infectious pneumonia and/or infectious acute respiratory distress syndrome (ARDS), wherein the patient is or has been treated with an inhibitor of C5a activity.
3. The inhibitor of C5a activity or the inhibitor of IL-6 activity for use according to claim 1 or 2, wherein the inhibitor of IL-6 activity binds specifically to IL-6 or the IL-6 receptor (IL-6R), preferably comprises a protein ligand.
4. The inhibitor of C5a activity or the inhibitor of IL-6 activity for use according to any one of the previous claims, wherein the inhibitor of C5a activity binds specifically to C5a or to the C5a receptor (C5aR), preferably comprises:
(i) a protein ligand specifically binding to C5a or the C5aR, preferably specifically binding to C5a
(ii) a small molecule C5aR antagonist, preferably according to general formula (I).
5. The inhibitor of C5a activity or the inhibitor of IL-6 activity for use according to claim
3 or 4, wherein the protein ligand is:
(i) an antibody or an antigen binding fragment thereof, preferably the protein ligand comprises a VH and a VL domain of an antibody, a Fab fragment, a Fab’ fragment, a heavy chain antibody, a single-domain antibody (sdAb), variable domain of a heavy chain antibody, a VHH, a nanobody, a single-chain variable fragment (scFv), a tandem scFv, or a single-chain diabody;
(ii) an antibody-like protein, preferably an affibody, anticalin, or an designed ankyrin repeat protein;
(iii) an inhibitory variant of C5a; or
(iv) an inhibitory variant of C5a receptor.
6. The inhibitor of C5a activity or the inhibitor of IL-6 activity for use according to claim 4 or 5 , wherein the protein ligand specifically binding to C5a binds to a conformational epitope of C5a formed by amino acid sequences NDETCEQRA (SEQ ID NO: 2) and SHKDMQL (SEQ ID NO: 3), and binds to at least one amino acid within the amino acid sequence according to SEQ ID NO: 2 and to at least one amino acid within the amino acid sequence according to SEQ ID NO: 3, wherein the specific binding to the amino acids of the conformational epitope is determined by alanine scanning.
7. The inhibitor of C5a activity or the inhibitor of IL-6 activity for use according to claim 6, wherein the protein ligand specifically binding to C5a is an antibody or antigenbinding fragment thereof that: binds to at least one amino acid of an amino acid sequence according to DETCEQR (SEQ ID NO: 4); and/or binds to at least one amino acid of an amino acid sequence according to HKDMQ (SEQ ID NO: 5), preferably to KDM.
8. The inhibitor of C5a activity or the inhibitor of IL-6 activity for use according to any one of claims 4 to 7, wherein the protein ligand specifically binding to C5a is an antibody or antigen-binding fragment thereof comprising a variable domain of a heavy chain (VH) and a variable domain of a light chain (VL), wherein the VL comprises:
(i) a light chain CDR3 sequence as set forth in SEQ ID NO: 6; or
(ii) a light chain CDR3 sequence as set forth in SEQ ID NO: 7; wherein said antibody or antigen-binding fragment thereof has a binding constant to C5a with a Ka value of 10 nM or less and exhibits at least 80% blocking activity for a biological effect induced by C5a, optionally the VL further comprises a FR2 region as set forth in SEQ ID NO: 8 or SEQ ID NO: 39.
9. The inhibitor of C5a activity or the inhibitor of IL-6 activity for use according to claim 8, wherein the protein ligand specifically binding to C5a further comprises at least one of the following sequences:
(i) a heavy chain CDR2 sequence according to SEQ ID NO: 9;
(ii) a heavy chain CDR2 sequence according to SEQ ID NO: 10;
(iii) a light chain CDR2 sequence according to SEQ ID NO: 11;
(iv) a light chain CDR2 sequence according to SEQ ID NO: 12;
(v) a heavy chain CDR1 sequence according to SEQ ID NO: 13;
(vi) a heavy chain CDR1 sequence according to SEQ ID NO: 14;
(vii) a light chain CDR1 sequence according to SEQ ID NO: 15;
(viii) a light chain CDR1 sequence according to SEQ ID NO: 16;
(ix) a heavy chain CDR3 sequence according to SEQ ID NO: 17; or
(x) a heavy chain CDR3 sequence according to SEQ ID NO: 18.
10. The inhibitor of C5a activity or the inhibitor of IL-6 activity for use according to any one of claims 4 to 9, wherein the protein ligand specifically binding to C5a comprises:
(i) a light chain CDR3 sequence according to SEQ ID NO: 6, a light chain CDR2 sequence according to SEQ ID NO: 11, a light chain CDR1 sequence according to SEQ ID NO: 15, a heavy chain CDR3 sequence according to SEQ ID NO: 17, a heavy chain CDR2 sequence according to SEQ ID NO: 9, and a heavy chain CDR1 sequence according to SEQ ID NO: 13; or
(ii) a light chain CDR3 sequence according to SEQ ID NO: 7, a light chain CDR2 sequence according to SEQ ID NO: 12, and a light chain CDR1 sequence according to SEQ ID NO: 16, a heavy chain CDR3 sequence according to SEQ ID NO: 18, a heavy chain CDR2 sequence according to SEQ ID NO: 10, and a heavy chain CDR1 sequence according to SEQ ID NO: 14.
11. The inhibitor of C5a activity or the inhibitor of IL-6 activity for use according to any one of claims 4 to 10, wherein the protein ligand specifically binding to C5a is selected from the group consisting of:
(a) IFX-1 (vilobelimab), IFX-2, INab708, BNJ364, BNJ367, BNJ371, BNJ378, BNJ366, BNJ369, BNJ381, BNJ383, MEDL7814 or ALXN-1007, or an antigen-binding fragment thereof; or
(b) an antibody or an antigen-binding fragment thereof, wherein said antibody or antigen-binding fragment thereof competes with one of the antibodies indicated under (a) for binding to C5a.
12. The inhibitor of C5a activity or the inhibitor of IL-6 activity for use according to any one of claims 3 to 11, wherein:
(a) the protein ligand specifically binding to IL-6R is selected from Tocilizumab, Sarilumab, Levilimab, and Vobarilizumab; more preferably wherein the inhibitor of IL-6 activity is Tocilizumab; or
(b) the protein ligand specifically binding to IL-6 is selected from Siltuximab, Sirukumab, Olokizumab, Elsilimomab, Clazakinumab, PF-423691 and EBI- 031.
13. The inhibitor of C5a activity or the inhibitor of IL-6 activity for use according to any one of the previous claims, wherein
(a) the infectious pneumonia is selected from viral pneumonia, fungal pneumonia or bacterial pneumonia, preferably CO VID-19 pneumonia; and/or
(b) the infectious ARDS is selected from viral ARDS and bacterial ARDS; preferably COVID-19 ARDS.
14. The inhibitor of C5a activity or the inhibitor of IL-6 activity for use according to any one of the previous claims, wherein the inhibitor of C5a activity is vilobelimab and the inhibitor of IL-6 activity is tocilizumab.
15. The inhibitor of C5a activity or the inhibitor of IL-6 activity for use according to any one of the previous claims, for administration in a regime comprising an induction phase followed by a maintenance phase, wherein the inhibitor of C5a activity is administered in the induction phase and the maintenance phase and the inhibitor of IL-6 activity is administered:
(I) within 0-30 days prior to the induction phase,
(II) during the induction phase, and/or
(III) during the maintenance phase.
16. The inhibitor of C5a activity and the inhibitor of IL-6 activity for use according to claim 15, wherein
(i) the induction phase comprises or essentially consists of a period of 1 to 10 days, preferably 2 to 8 days, more preferably 7-8 days, wherein 1-8 doses, preferably 2-4 doses, comprising each 200-1600 mg, preferably 400-1200 mg of said inhibitor of C5a activity, are administered; and
(ii) the maintenance phase comprises or essentially consists of a period of at least 2 weeks, preferably 2-4 weeks, wherein a dose, comprising 200-1600 mg, preferably 400-1200 mg of said inhibitor of C5a activity, is administered every 1-4 weeks, preferably every week; preferably the inhibitor of IL-6 activity is administered at a weight dependent dose of 8mg/kg for patients at or above 30 kg weight or 12 mg/kg for patients of less than 30 kg weight.
17. The inhibitor of C5a activity and the inhibitor of IL-6 activity for use according to any one of claims 15 and 16, wherein
(i) 1 or 2 doses (preferably single dose) of the inhibitor of IL-6 activity (preferably tocilizumab) is administered within 0 to 14 days prior and/or after the start of the induction phase (preferably within 0 to 7 days prior or after the start of the induction phase); and/or
(ii) the inhibitor of IL-6 activity (preferably tocilizumab) is administered at a weight dependent dose of 8mg/kg for patients at or above 30 kg weight or 12 mg/kg for patients of less than 30 kg weight.
18. A kit of parts comprising an inhibitor of C5a activity, preferably vilobelimab, and an inhibitor of IL-6 activity, preferably tocilizumab.
Priority Applications (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
PCT/EP2023/064290 WO2024245527A1 (en) | 2023-05-26 | 2023-05-26 | Treatment of pneumonia and ards with inhibitors of c5a and il-6 activity |
Applications Claiming Priority (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
PCT/EP2023/064290 WO2024245527A1 (en) | 2023-05-26 | 2023-05-26 | Treatment of pneumonia and ards with inhibitors of c5a and il-6 activity |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2024245527A1 true WO2024245527A1 (en) | 2024-12-05 |
Family
ID=86732770
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/EP2023/064290 WO2024245527A1 (en) | 2023-05-26 | 2023-05-26 | Treatment of pneumonia and ards with inhibitors of c5a and il-6 activity |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2024245527A1 (en) |
Citations (8)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US5939598A (en) | 1990-01-12 | 1999-08-17 | Abgenix, Inc. | Method of making transgenic mice lacking endogenous heavy chains |
US20030118592A1 (en) | 2001-01-17 | 2003-06-26 | Genecraft, Inc. | Binding domain-immunoglobulin fusion proteins |
US20030133939A1 (en) | 2001-01-17 | 2003-07-17 | Genecraft, Inc. | Binding domain-immunoglobulin fusion proteins |
WO2011063980A1 (en) | 2009-11-26 | 2011-06-03 | Inflarx Gmbh | Anti-c5a binding moieties with high blocking activity |
WO2011137395A1 (en) | 2010-04-30 | 2011-11-03 | Rother Russell P | Anti-c5a antibodies and methods for using the antibodies |
WO2020182384A1 (en) | 2019-03-11 | 2020-09-17 | Inflarx Gmbh | Fused piperidinyl bicyclic and related compounds as modulators of c5a receptor |
WO2021188601A1 (en) * | 2020-03-16 | 2021-09-23 | University Of Southern California | Methods to prevent, ameliorate and treat complications from viral infections |
WO2021190770A1 (en) | 2020-03-27 | 2021-09-30 | Inflarx Gmbh | INHIBITORS OF C5A FOR THE TREATMENT OF CORONA VIRUS Infection |
-
2023
- 2023-05-26 WO PCT/EP2023/064290 patent/WO2024245527A1/en unknown
Patent Citations (9)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US5939598A (en) | 1990-01-12 | 1999-08-17 | Abgenix, Inc. | Method of making transgenic mice lacking endogenous heavy chains |
US20030118592A1 (en) | 2001-01-17 | 2003-06-26 | Genecraft, Inc. | Binding domain-immunoglobulin fusion proteins |
US20030133939A1 (en) | 2001-01-17 | 2003-07-17 | Genecraft, Inc. | Binding domain-immunoglobulin fusion proteins |
WO2011063980A1 (en) | 2009-11-26 | 2011-06-03 | Inflarx Gmbh | Anti-c5a binding moieties with high blocking activity |
US20120231008A1 (en) | 2009-11-26 | 2012-09-13 | Renfeng Guo | Anti-C5A Binding Moieties with High Blocking Activity |
WO2011137395A1 (en) | 2010-04-30 | 2011-11-03 | Rother Russell P | Anti-c5a antibodies and methods for using the antibodies |
WO2020182384A1 (en) | 2019-03-11 | 2020-09-17 | Inflarx Gmbh | Fused piperidinyl bicyclic and related compounds as modulators of c5a receptor |
WO2021188601A1 (en) * | 2020-03-16 | 2021-09-23 | University Of Southern California | Methods to prevent, ameliorate and treat complications from viral infections |
WO2021190770A1 (en) | 2020-03-27 | 2021-09-30 | Inflarx Gmbh | INHIBITORS OF C5A FOR THE TREATMENT OF CORONA VIRUS Infection |
Non-Patent Citations (22)
Title |
---|
"A multilingual glossary of biotechnological terms: (IUPAC Recommendations", 1995, HELVETICA CHIMICA ACTA |
"UniProtKB", Database accession no. P05231 |
ALMAGROFRANSSON, FRONTIERS IN BIOSCIENCE, vol. 13, 2008, pages 1619 - 1633 |
ANNANE DJILLALI ET AL: "Intravenous ravulizumab in mechanically ventilated patients hospitalised with severe COVID-19: a phase 3, multicentre, open-label, randomised controlled trial", THE LANCET. RESPIRATORY MEDICINE, vol. 11, no. 12, 20 March 2023 (2023-03-20), Oxford, pages 1051 - 1063, XP093109282, ISSN: 2213-2600, DOI: 10.1016/S2213-2600(23)00082-6 * |
ANONYMOUS: "Emergency Use Authorization (EUA) for Vilobelimab (IFX-1) Center for Drug Evaluation and Research (CDER) Review", 1 February 2023 (2023-02-01), XP093109280, Retrieved from the Internet <URL:https://www.gohibic.com/> [retrieved on 20231205] * |
BINZ H.K. ET AL.: "Engineering novel binding proteins from non-immunoglobulin domains", NAT. BIOTECHNOL., vol. 23, no. 10, 2005, pages 1257 - 1268 |
BIRD ET AL., SCIENCE, vol. 242, 1988, pages 423 - 426 |
CHOTHIA ET AL., J. MOL. BIOL., vol. 196, 1987, pages 901 - 917 |
HEAP C.J. ET AL., J. GEN. VIROL., vol. 86, 2005, pages 1791 - 1800 |
HOLLIGER P. ET AL., PROC. NATL. ACAD. SCI. U.S.A., vol. 90, no. 14, 1993, pages 6444 - 6448 |
HOPKEN U ET AL., EUR J IMMUNOL, vol. 26, no. 5, 1996, pages 1103 - 1109 |
HUSTON ET AL., PROC. NATL. ACAD. SCI. USA, vol. 85, 1988, pages 5879 - 5883 |
KABAT ET AL., J. BIOL. CHEM., vol. 252, 1977, pages 6609 - 6616 |
KABAT ET AL.: "Sequences of proteins of immunological interest", 1991, U.S. DEPT. OF HEALTH AND HUMAN SERVICES |
MACCALLUM ET AL., J. MOL. BIOL., vol. 262, 1996, pages 732 - 745 |
MOURVILLIER BRUNO ET AL: "LB1529. Randomized, Controlled Phase 3 Study of anti-C5a Vilobelimab's Effect on Mortality in Critically Ill COVID-19 Patients: A Therapy for Viral Pneumonia", OPEN FORUM INFECTIOUS DISEASES, vol. 9, no. Supplement_2, 15 December 2022 (2022-12-15), XP093109121, ISSN: 2328-8957, DOI: 10.1093/ofid/ofac492.1875 * |
QIU X.Q, NATURE BIOTECHNOLOGY, vol. 25, no. 8, 2007, pages 921 - 929 |
RIEDEMANN NC ET AL., J IMMUNOL, vol. 173, no. 2, 2004, pages 1355 - 1359 |
RIEDEMANN NC ET AL., J. IMMUNOL., vol. 168, no. 4, 2002, pages 1919 - 1925 |
RITTIRSCH D ET AL., NAT REV IMMUNOL, vol. 8, no. 10, 2008, pages 776 - 787 |
STRIETER RM ET AL., AM J PATHOL, vol. 141, no. 2, 1992, pages 397 - 407 |
WARD ET AL., NATURE, vol. 341, pages 544 - 546 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
US10336831B2 (en) | Use of anti-endoglin antibodies for treating ocular fibrosis | |
JP7324565B2 (en) | Antibodies against CD127 | |
CN105916519B (en) | Anti-Siglec-8 antibody and method of use | |
RU2731713C2 (en) | Use of il-18-binding protein (il-18bp) in inflammatory diseases | |
US10858425B2 (en) | Pan-ELR+ CXC chemokine antibodies | |
US9926375B2 (en) | Anti-endoglin antibodies and uses thereof | |
JP6105146B2 (en) | Pan-ELR + CXC chemokine antibody | |
US11845802B2 (en) | Combination therapy with a glucagon receptor (GCGR) antibody and an anti-CD3 antibody | |
EP2279001B1 (en) | Inhibitors of gm-csf and il-17 for therapy | |
EP2767549A1 (en) | Anti-CD26 antibodies and uses thereof | |
TW201348255A (en) | Anti-big-endothelin-1 (big-ET-1) antibodies and uses thereof | |
JP2021113190A (en) | Anti-cd40 antibodies for use in prevention of graft rejection | |
US12180274B2 (en) | Treatment of pneumonia and ARDS with inhibitors of C5a and IL-6 activity | |
WO2024245527A1 (en) | Treatment of pneumonia and ards with inhibitors of c5a and il-6 activity | |
US20230272087A1 (en) | Method of treating or preventing acute respiratory distress syndrome | |
CA2940869A1 (en) | Antibodies to matrix metalloproteinase 9 and methods of use thereof | |
JP2021500865A (en) | Staphylococcus bispecific antigen-binding molecule that binds to target antigens and complement components and their use | |
CA3236006A1 (en) | Methods and compositions for treating systemic lupus erythematosus (sle) with mosunetuzumab | |
AU2021237566A1 (en) | Anti-glycan antibodies and uses thereof | |
US20230416381A1 (en) | Methods for treating or preventing acute respiratory distress syndrome | |
US20220213210A1 (en) | Combination therapy of multiple sclerosis comprising a cd20 ligand |
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: 23729720 Country of ref document: EP Kind code of ref document: A1 |