WO2022212914A1 - Methods of treating viral diseases - Google Patents
Methods of treating viral diseases Download PDFInfo
- Publication number
- WO2022212914A1 WO2022212914A1 PCT/US2022/023162 US2022023162W WO2022212914A1 WO 2022212914 A1 WO2022212914 A1 WO 2022212914A1 US 2022023162 W US2022023162 W US 2022023162W WO 2022212914 A1 WO2022212914 A1 WO 2022212914A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- virus
- combinations
- drug
- pharmaceutical composition
- patient
- Prior art date
Links
- 238000000034 method Methods 0.000 title claims abstract description 130
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 title claims abstract description 75
- 201000010099 disease Diseases 0.000 title claims abstract description 70
- 230000003612 virological effect Effects 0.000 title claims description 39
- RVLCUCVJZVRNDC-IMJSIDKUSA-N 2-[(2s,5s)-5-methyl-3,6-dioxopiperazin-2-yl]acetic acid Chemical compound C[C@@H]1NC(=O)[C@H](CC(O)=O)NC1=O RVLCUCVJZVRNDC-IMJSIDKUSA-N 0.000 claims abstract description 110
- 108010038239 aspartyl-alanyl-diketopiperazine Proteins 0.000 claims abstract description 110
- 239000000203 mixture Substances 0.000 claims abstract description 75
- 239000008194 pharmaceutical composition Substances 0.000 claims abstract description 74
- 108091006905 Human Serum Albumin Proteins 0.000 claims abstract description 31
- 102000008100 Human Serum Albumin Human genes 0.000 claims abstract description 31
- 230000001404 mediated effect Effects 0.000 claims abstract description 26
- 102000009027 Albumins Human genes 0.000 claims abstract description 20
- 108010088751 Albumins Proteins 0.000 claims abstract description 20
- 239000003814 drug Substances 0.000 claims description 69
- 229940079593 drug Drugs 0.000 claims description 57
- 241000700605 Viruses Species 0.000 claims description 40
- 206010001052 Acute respiratory distress syndrome Diseases 0.000 claims description 37
- WWZKQHOCKIZLMA-UHFFFAOYSA-N octanoic acid Chemical compound CCCCCCCC(O)=O WWZKQHOCKIZLMA-UHFFFAOYSA-N 0.000 claims description 36
- DZTHIGRZJZPRDV-LBPRGKRZSA-N N-acetyl-L-tryptophan Chemical compound C1=CC=C2C(C[C@H](NC(=O)C)C(O)=O)=CNC2=C1 DZTHIGRZJZPRDV-LBPRGKRZSA-N 0.000 claims description 34
- DZTHIGRZJZPRDV-UHFFFAOYSA-N Nalpha-Acetyltryptophan Natural products C1=CC=C2C(CC(NC(=O)C)C(O)=O)=CNC2=C1 DZTHIGRZJZPRDV-UHFFFAOYSA-N 0.000 claims description 34
- 229940116191 n-acetyltryptophan Drugs 0.000 claims description 34
- 206010061218 Inflammation Diseases 0.000 claims description 32
- 230000004054 inflammatory process Effects 0.000 claims description 32
- 208000013616 Respiratory Distress Syndrome Diseases 0.000 claims description 31
- 201000000028 adult respiratory distress syndrome Diseases 0.000 claims description 31
- 210000004072 lung Anatomy 0.000 claims description 28
- 208000024891 symptom Diseases 0.000 claims description 24
- 241000711573 Coronaviridae Species 0.000 claims description 19
- 239000005635 Caprylic acid (CAS 124-07-2) Substances 0.000 claims description 18
- QVGXLLKOCUKJST-UHFFFAOYSA-N atomic oxygen Chemical compound [O] QVGXLLKOCUKJST-UHFFFAOYSA-N 0.000 claims description 18
- WWZKQHOCKIZLMA-UHFFFAOYSA-M octanoate Chemical compound CCCCCCCC([O-])=O WWZKQHOCKIZLMA-UHFFFAOYSA-M 0.000 claims description 18
- 229960002446 octanoic acid Drugs 0.000 claims description 18
- 239000001301 oxygen Substances 0.000 claims description 18
- 229910052760 oxygen Inorganic materials 0.000 claims description 18
- 208000036142 Viral infection Diseases 0.000 claims description 17
- 208000015181 infectious disease Diseases 0.000 claims description 17
- 230000009385 viral infection Effects 0.000 claims description 17
- 208000002606 Paramyxoviridae Infections Diseases 0.000 claims description 16
- 230000000241 respiratory effect Effects 0.000 claims description 16
- 101000800483 Homo sapiens Toll-like receptor 8 Proteins 0.000 claims description 13
- 102100033110 Toll-like receptor 8 Human genes 0.000 claims description 13
- 206010069351 acute lung injury Diseases 0.000 claims description 12
- 241000725643 Respiratory syncytial virus Species 0.000 claims description 11
- LIENCHBZNNMNKG-OJFNHCPVSA-N nirmatrelvir Chemical compound CC1([C@@H]2[C@H]1[C@H](N(C2)C(=O)[C@H](C(C)(C)C)NC(=O)C(F)(F)F)C(=O)N[C@@H](C[C@@H]3CCNC3=O)C#N)C LIENCHBZNNMNKG-OJFNHCPVSA-N 0.000 claims description 11
- -1 L - hydroxycytidine Chemical compound 0.000 claims description 10
- 230000000840 anti-viral effect Effects 0.000 claims description 10
- JYGXADMDTFJGBT-VWUMJDOOSA-N hydrocortisone Chemical compound O=C1CC[C@]2(C)[C@H]3[C@@H](O)C[C@](C)([C@@](CC4)(O)C(=O)CO)[C@@H]4[C@@H]3CCC2=C1 JYGXADMDTFJGBT-VWUMJDOOSA-N 0.000 claims description 10
- 229940002612 prodrug Drugs 0.000 claims description 10
- 239000000651 prodrug Substances 0.000 claims description 10
- 208000025721 COVID-19 Diseases 0.000 claims description 9
- 241000531123 GB virus C Species 0.000 claims description 9
- 206010035664 Pneumonia Diseases 0.000 claims description 9
- 239000003246 corticosteroid Substances 0.000 claims description 9
- 230000002829 reductive effect Effects 0.000 claims description 9
- 241000712461 unidentified influenza virus Species 0.000 claims description 9
- 241000709661 Enterovirus Species 0.000 claims description 8
- 229960003957 dexamethasone Drugs 0.000 claims description 8
- 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 claims description 8
- 150000003833 nucleoside derivatives Chemical class 0.000 claims description 8
- 150000003839 salts Chemical class 0.000 claims description 8
- 208000005777 Lupus Nephritis Diseases 0.000 claims description 7
- 201000003176 Severe Acute Respiratory Syndrome Diseases 0.000 claims description 7
- 239000004012 Tofacitinib Substances 0.000 claims description 7
- 229950000971 baricitinib Drugs 0.000 claims description 7
- XUZMWHLSFXCVMG-UHFFFAOYSA-N baricitinib Chemical compound C1N(S(=O)(=O)CC)CC1(CC#N)N1N=CC(C=2C=3C=CNC=3N=CN=2)=C1 XUZMWHLSFXCVMG-UHFFFAOYSA-N 0.000 claims description 7
- 210000004369 blood Anatomy 0.000 claims description 7
- 239000008280 blood Substances 0.000 claims description 7
- 210000002216 heart Anatomy 0.000 claims description 7
- 206010025135 lupus erythematosus Diseases 0.000 claims description 7
- 230000003472 neutralizing effect Effects 0.000 claims description 7
- 239000002777 nucleoside Substances 0.000 claims description 7
- 239000000137 peptide hydrolase inhibitor Substances 0.000 claims description 7
- RWWYLEGWBNMMLJ-MEUHYHILSA-N remdesivir Drugs C([C@@H]1[C@H]([C@@H](O)[C@@](C#N)(O1)C=1N2N=CN=C(N)C2=CC=1)O)OP(=O)(N[C@@H](C)C(=O)OCC(CC)CC)OC1=CC=CC=C1 RWWYLEGWBNMMLJ-MEUHYHILSA-N 0.000 claims description 7
- RWWYLEGWBNMMLJ-YSOARWBDSA-N remdesivir Chemical compound NC1=NC=NN2C1=CC=C2[C@]1([C@@H]([C@@H]([C@H](O1)CO[P@](=O)(OC1=CC=CC=C1)N[C@H](C(=O)OCC(CC)CC)C)O)O)C#N RWWYLEGWBNMMLJ-YSOARWBDSA-N 0.000 claims description 7
- 208000011580 syndromic disease Diseases 0.000 claims description 7
- 210000001519 tissue Anatomy 0.000 claims description 7
- 229960001350 tofacitinib Drugs 0.000 claims description 7
- UJLAWZDWDVHWOW-YPMHNXCESA-N tofacitinib Chemical compound C[C@@H]1CCN(C(=O)CC#N)C[C@@H]1N(C)C1=NC=NC2=C1C=CN2 UJLAWZDWDVHWOW-YPMHNXCESA-N 0.000 claims description 7
- 238000000108 ultra-filtration Methods 0.000 claims description 7
- 229940125678 AZD7442 Drugs 0.000 claims description 6
- 208000009304 Acute Kidney Injury Diseases 0.000 claims description 6
- 241001678559 COVID-19 virus Species 0.000 claims description 6
- 206010013975 Dyspnoeas Diseases 0.000 claims description 6
- 241000712431 Influenza A virus Species 0.000 claims description 6
- 241000713196 Influenza B virus Species 0.000 claims description 6
- 208000033626 Renal failure acute Diseases 0.000 claims description 6
- 240000001068 Thogoto virus Species 0.000 claims description 6
- 208000004006 Tick-borne encephalitis Diseases 0.000 claims description 6
- 201000011040 acute kidney failure Diseases 0.000 claims description 6
- 210000003734 kidney Anatomy 0.000 claims description 6
- 229940125674 nirmatrelvir Drugs 0.000 claims description 6
- 229950006348 sarilumab Drugs 0.000 claims description 6
- 230000000153 supplemental effect Effects 0.000 claims description 6
- 229960003989 tocilizumab Drugs 0.000 claims description 6
- 206010008469 Chest discomfort Diseases 0.000 claims description 5
- 208000029523 Interstitial Lung disease Diseases 0.000 claims description 5
- FQISKWAFAHGMGT-SGJOWKDISA-M Methylprednisolone sodium succinate Chemical compound [Na+].C([C@@]12C)=CC(=O)C=C1[C@@H](C)C[C@@H]1[C@@H]2[C@@H](O)C[C@]2(C)[C@@](O)(C(=O)COC(=O)CCC([O-])=O)CC[C@H]21 FQISKWAFAHGMGT-SGJOWKDISA-M 0.000 claims description 5
- 208000025370 Middle East respiratory syndrome Diseases 0.000 claims description 5
- 241000127282 Middle East respiratory syndrome-related coronavirus Species 0.000 claims description 5
- 229940124158 Protease/peptidase inhibitor Drugs 0.000 claims description 5
- 206010038687 Respiratory distress Diseases 0.000 claims description 5
- NCDNCNXCDXHOMX-UHFFFAOYSA-N Ritonavir Natural products C=1C=CC=CC=1CC(NC(=O)OCC=1SC=NC=1)C(O)CC(CC=1C=CC=CC=1)NC(=O)C(C(C)C)NC(=O)N(C)CC1=CSC(C(C)C)=N1 NCDNCNXCDXHOMX-UHFFFAOYSA-N 0.000 claims description 5
- 230000010100 anticoagulation Effects 0.000 claims description 5
- 229940051183 casirivimab Drugs 0.000 claims description 5
- 229940051243 etesevimab Drugs 0.000 claims description 5
- ZCGNOVWYSGBHAU-UHFFFAOYSA-N favipiravir Chemical compound NC(=O)C1=NC(F)=CNC1=O ZCGNOVWYSGBHAU-UHFFFAOYSA-N 0.000 claims description 5
- 229950008454 favipiravir Drugs 0.000 claims description 5
- 239000000706 filtrate Substances 0.000 claims description 5
- 229960004038 fluvoxamine Drugs 0.000 claims description 5
- CJOFXWAVKWHTFT-XSFVSMFZSA-N fluvoxamine Chemical compound COCCCC\C(=N/OCCN)C1=CC=C(C(F)(F)F)C=C1 CJOFXWAVKWHTFT-XSFVSMFZSA-N 0.000 claims description 5
- 229960000890 hydrocortisone Drugs 0.000 claims description 5
- 229940051184 imdevimab Drugs 0.000 claims description 5
- 229950007439 lenzilumab Drugs 0.000 claims description 5
- 229960004584 methylprednisolone Drugs 0.000 claims description 5
- HTNPEHXGEKVIHG-ZJTJHKMLSA-N molnupiravir Chemical compound CC(C)C(=O)OC[C@H]1O[C@H](C(O)C1O)N1C=C\C(NC1=O)=N\O HTNPEHXGEKVIHG-ZJTJHKMLSA-N 0.000 claims description 5
- 229940075124 molnupiravir Drugs 0.000 claims description 5
- 229940125675 paxlovid Drugs 0.000 claims description 5
- 229960004618 prednisone Drugs 0.000 claims description 5
- XOFYZVNMUHMLCC-ZPOLXVRWSA-N prednisone Chemical compound O=C1C=C[C@]2(C)[C@H]3C(=O)C[C@](C)([C@@](CC4)(O)C(=O)CO)[C@@H]4[C@@H]3CCC2=C1 XOFYZVNMUHMLCC-ZPOLXVRWSA-N 0.000 claims description 5
- 229960000311 ritonavir Drugs 0.000 claims description 5
- NCDNCNXCDXHOMX-XGKFQTDJSA-N ritonavir Chemical compound N([C@@H](C(C)C)C(=O)N[C@H](C[C@H](O)[C@H](CC=1C=CC=CC=1)NC(=O)OCC=1SC=NC=1)CC=1C=CC=CC=1)C(=O)N(C)CC1=CSC(C(C)C)=N1 NCDNCNXCDXHOMX-XGKFQTDJSA-N 0.000 claims description 5
- 239000001226 triphosphate Substances 0.000 claims description 5
- 206010011224 Cough Diseases 0.000 claims description 4
- 241000608297 Getah virus Species 0.000 claims description 4
- 241000342334 Human metapneumovirus Species 0.000 claims description 4
- 206010037660 Pyrexia Diseases 0.000 claims description 4
- 102100038081 Signal transducer CD24 Human genes 0.000 claims description 4
- 230000001430 anti-depressive effect Effects 0.000 claims description 4
- 239000000935 antidepressant agent Substances 0.000 claims description 4
- 241001493065 dsRNA viruses Species 0.000 claims description 4
- 230000036541 health Effects 0.000 claims description 4
- 241000710929 Alphavirus Species 0.000 claims description 3
- 241000712892 Arenaviridae Species 0.000 claims description 3
- 241000712891 Arenavirus Species 0.000 claims description 3
- 241001533362 Astroviridae Species 0.000 claims description 3
- 241000711404 Avian avulavirus 1 Species 0.000 claims description 3
- 241000035315 Avulavirus Species 0.000 claims description 3
- 241000710946 Barmah Forest virus Species 0.000 claims description 3
- 241001493154 Bunyamwera virus Species 0.000 claims description 3
- 241000714198 Caliciviridae Species 0.000 claims description 3
- 201000009182 Chikungunya Diseases 0.000 claims description 3
- 241000709687 Coxsackievirus Species 0.000 claims description 3
- 241001520695 Duvenhage lyssavirus Species 0.000 claims description 3
- 208000000059 Dyspnea Diseases 0.000 claims description 3
- 241000710945 Eastern equine encephalitis virus Species 0.000 claims description 3
- 241001466953 Echovirus Species 0.000 claims description 3
- 206010014596 Encephalitis Japanese B Diseases 0.000 claims description 3
- 241000991587 Enterovirus C Species 0.000 claims description 3
- 206010066919 Epidemic polyarthritis Diseases 0.000 claims description 3
- 241000710781 Flaviviridae Species 0.000 claims description 3
- 241000710831 Flavivirus Species 0.000 claims description 3
- 241000150562 Hantaan orthohantavirus Species 0.000 claims description 3
- 241000893570 Hendra henipavirus Species 0.000 claims description 3
- 241000035314 Henipavirus Species 0.000 claims description 3
- 241000711557 Hepacivirus Species 0.000 claims description 3
- 208000005176 Hepatitis C Diseases 0.000 claims description 3
- 241000709715 Hepatovirus Species 0.000 claims description 3
- 241001122120 Hepeviridae Species 0.000 claims description 3
- 101000884271 Homo sapiens Signal transducer CD24 Proteins 0.000 claims description 3
- 241000598436 Human T-cell lymphotropic virus Species 0.000 claims description 3
- 241000598171 Human adenovirus sp. Species 0.000 claims description 3
- 241000725303 Human immunodeficiency virus Species 0.000 claims description 3
- 201000005807 Japanese encephalitis Diseases 0.000 claims description 3
- 208000003140 Kyasanur forest disease Diseases 0.000 claims description 3
- 241000713666 Lentivirus Species 0.000 claims description 3
- 206010024887 Louping ill Diseases 0.000 claims description 3
- 241000712899 Lymphocytic choriomeningitis mammarenavirus Species 0.000 claims description 3
- 241000711828 Lyssavirus Species 0.000 claims description 3
- 241000608292 Mayaro virus Species 0.000 claims description 3
- 241000712079 Measles morbillivirus Species 0.000 claims description 3
- 241000351643 Metapneumovirus Species 0.000 claims description 3
- 241000711513 Mononegavirales Species 0.000 claims description 3
- 241000712045 Morbillivirus Species 0.000 claims description 3
- 241000868135 Mucambo virus Species 0.000 claims description 3
- 241000711386 Mumps virus Species 0.000 claims description 3
- 241000710908 Murray Valley encephalitis virus Species 0.000 claims description 3
- 208000000112 Myalgia Diseases 0.000 claims description 3
- 241001457453 Nairobi sheep disease virus Species 0.000 claims description 3
- 241000526636 Nipah henipavirus Species 0.000 claims description 3
- 241001263478 Norovirus Species 0.000 claims description 3
- 241000713112 Orthobunyavirus Species 0.000 claims description 3
- 241000150452 Orthohantavirus Species 0.000 claims description 3
- 241000712464 Orthomyxoviridae Species 0.000 claims description 3
- 241000150218 Orthonairovirus Species 0.000 claims description 3
- 241000150350 Peribunyaviridae Species 0.000 claims description 3
- 241000713137 Phlebovirus Species 0.000 claims description 3
- 241000709664 Picornaviridae Species 0.000 claims description 3
- 241000711902 Pneumovirus Species 0.000 claims description 3
- 241000711798 Rabies lyssavirus Species 0.000 claims description 3
- 241000712907 Retroviridae Species 0.000 claims description 3
- 241000711931 Rhabdoviridae Species 0.000 claims description 3
- 241001325464 Rhinovirus A Species 0.000 claims description 3
- 241001325459 Rhinovirus B Species 0.000 claims description 3
- 241000907332 Rocio virus Species 0.000 claims description 3
- 241000710942 Ross River virus Species 0.000 claims description 3
- 241000710799 Rubella virus Species 0.000 claims description 3
- 241000710801 Rubivirus Species 0.000 claims description 3
- 241001533467 Rubulavirus Species 0.000 claims description 3
- 208000032108 Russian spring-summer encephalitis Diseases 0.000 claims description 3
- 241000608282 Sagiyama virus Species 0.000 claims description 3
- 241001135555 Sandfly fever Sicilian virus Species 0.000 claims description 3
- 241000369753 Sapporo virus Species 0.000 claims description 3
- 241000710961 Semliki Forest virus Species 0.000 claims description 3
- 241000710960 Sindbis virus Species 0.000 claims description 3
- 241000907333 Spondweni virus Species 0.000 claims description 3
- 241000710924 Togaviridae Species 0.000 claims description 3
- 241000868137 Tonate virus Species 0.000 claims description 3
- 241000711517 Torovirus Species 0.000 claims description 3
- 241000710959 Venezuelan equine encephalitis virus Species 0.000 claims description 3
- 241000710886 West Nile virus Species 0.000 claims description 3
- 241000710951 Western equine encephalitis virus Species 0.000 claims description 3
- 241000710772 Yellow fever virus Species 0.000 claims description 3
- 244000309743 astrovirus Species 0.000 claims description 3
- 210000004204 blood vessel Anatomy 0.000 claims description 3
- 210000004556 brain Anatomy 0.000 claims description 3
- 206010014599 encephalitis Diseases 0.000 claims description 3
- 206010016256 fatigue Diseases 0.000 claims description 3
- 208000005252 hepatitis A Diseases 0.000 claims description 3
- 201000010284 hepatitis E Diseases 0.000 claims description 3
- 239000012528 membrane Substances 0.000 claims description 3
- 208000013465 muscle pain Diseases 0.000 claims description 3
- 210000005036 nerve Anatomy 0.000 claims description 3
- 230000008520 organization Effects 0.000 claims description 3
- 238000001556 precipitation Methods 0.000 claims description 3
- 208000036273 reactive airway disease Diseases 0.000 claims description 3
- 208000013220 shortness of breath Diseases 0.000 claims description 3
- 238000000527 sonication Methods 0.000 claims description 3
- 238000000856 sucrose gradient centrifugation Methods 0.000 claims description 3
- 229940051021 yellow-fever virus Drugs 0.000 claims description 3
- 208000006820 Arthralgia Diseases 0.000 claims description 2
- 206010008479 Chest Pain Diseases 0.000 claims description 2
- 206010018364 Glomerulonephritis Diseases 0.000 claims description 2
- 206010027374 Mental impairment Diseases 0.000 claims description 2
- 208000010428 Muscle Weakness Diseases 0.000 claims description 2
- 206010028372 Muscular weakness Diseases 0.000 claims description 2
- XCUAIINAJCDIPM-XVFCMESISA-N N(4)-hydroxycytidine Chemical compound O[C@@H]1[C@H](O)[C@@H](CO)O[C@H]1N1C(=O)NC(=NO)C=C1 XCUAIINAJCDIPM-XVFCMESISA-N 0.000 claims description 2
- 206010068319 Oropharyngeal pain Diseases 0.000 claims description 2
- 206010033557 Palpitations Diseases 0.000 claims description 2
- 201000007100 Pharyngitis Diseases 0.000 claims description 2
- 208000036071 Rhinorrhea Diseases 0.000 claims description 2
- 206010039101 Rhinorrhoea Diseases 0.000 claims description 2
- 238000004587 chromatography analysis Methods 0.000 claims description 2
- 239000006199 nebulizer Substances 0.000 claims description 2
- 210000003491 skin Anatomy 0.000 claims description 2
- 241000701161 unidentified adenovirus Species 0.000 claims description 2
- 229940052143 bamlanivimab Drugs 0.000 claims 4
- 241000710827 Dengue virus 1 Species 0.000 claims 2
- 241000710815 Dengue virus 2 Species 0.000 claims 2
- 241000710872 Dengue virus 3 Species 0.000 claims 2
- 241000710844 Dengue virus 4 Species 0.000 claims 2
- 241001500351 Influenzavirus A Species 0.000 claims 2
- 241001500350 Influenzavirus B Species 0.000 claims 2
- 241001500343 Influenzavirus C Species 0.000 claims 2
- 241000401052 Influenzavirus D Species 0.000 claims 2
- 238000011260 co-administration Methods 0.000 claims 2
- 238000006213 oxygenation reaction Methods 0.000 claims 2
- 208000005069 pulmonary fibrosis Diseases 0.000 claims 2
- 230000009325 pulmonary function Effects 0.000 claims 2
- 208000027028 long COVID Diseases 0.000 claims 1
- 102000002689 Toll-like receptor Human genes 0.000 abstract description 17
- 108020000411 Toll-like receptor Proteins 0.000 abstract description 17
- BXRNXXXXHLBUKK-UHFFFAOYSA-N piperazine-2,5-dione Chemical compound O=C1CNC(=O)CN1 BXRNXXXXHLBUKK-UHFFFAOYSA-N 0.000 abstract description 6
- CKLJMWTZIZZHCS-REOHCLBHSA-N L-aspartic acid Chemical compound OC(=O)[C@@H](N)CC(O)=O CKLJMWTZIZZHCS-REOHCLBHSA-N 0.000 abstract description 3
- 150000001413 amino acids Chemical group 0.000 abstract description 3
- 235000003704 aspartic acid Nutrition 0.000 abstract description 3
- OQFSQFPPLPISGP-UHFFFAOYSA-N beta-carboxyaspartic acid Natural products OC(=O)C(N)C(C(O)=O)C(O)=O OQFSQFPPLPISGP-UHFFFAOYSA-N 0.000 abstract description 3
- QNAYBMKLOCPYGJ-REOHCLBHSA-N L-alanine Chemical compound C[C@H](N)C(O)=O QNAYBMKLOCPYGJ-REOHCLBHSA-N 0.000 abstract description 2
- 235000004279 alanine Nutrition 0.000 abstract description 2
- 238000011228 multimodal treatment Methods 0.000 abstract description 2
- 102100025248 C-X-C motif chemokine 10 Human genes 0.000 description 49
- 101000858088 Homo sapiens C-X-C motif chemokine 10 Proteins 0.000 description 48
- 238000011282 treatment Methods 0.000 description 37
- 210000004027 cell Anatomy 0.000 description 34
- 230000005764 inhibitory process Effects 0.000 description 26
- 102000004127 Cytokines Human genes 0.000 description 25
- 108090000695 Cytokines Proteins 0.000 description 25
- 208000023504 respiratory system disease Diseases 0.000 description 23
- 239000000243 solution Substances 0.000 description 23
- 101000669402 Homo sapiens Toll-like receptor 7 Proteins 0.000 description 22
- 102100039390 Toll-like receptor 7 Human genes 0.000 description 22
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 20
- 210000003819 peripheral blood mononuclear cell Anatomy 0.000 description 20
- XLCISDOVNFLSGO-VONOSFMSSA-N phorbol-12-myristate Chemical compound C([C@]1(O)C(=O)C(C)=C[C@H]1[C@@]1(O)[C@H](C)[C@H]2OC(=O)CCCCCCCCCCCCC)C(CO)=C[C@H]1[C@H]1[C@]2(O)C1(C)C XLCISDOVNFLSGO-VONOSFMSSA-N 0.000 description 19
- 102000019034 Chemokines Human genes 0.000 description 18
- 108010012236 Chemokines Proteins 0.000 description 18
- 206010052015 cytokine release syndrome Diseases 0.000 description 18
- 108010016626 Dipeptides Proteins 0.000 description 15
- 239000011780 sodium chloride Substances 0.000 description 15
- YXFVVABEGXRONW-UHFFFAOYSA-N Toluene Chemical compound CC1=CC=CC=C1 YXFVVABEGXRONW-UHFFFAOYSA-N 0.000 description 14
- 230000004913 activation Effects 0.000 description 13
- 150000001875 compounds Chemical class 0.000 description 12
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 11
- 206010050685 Cytokine storm Diseases 0.000 description 10
- 239000005557 antagonist Substances 0.000 description 10
- 230000037361 pathway Effects 0.000 description 10
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N Dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 9
- 230000015572 biosynthetic process Effects 0.000 description 9
- 230000000770 proinflammatory effect Effects 0.000 description 9
- 238000003786 synthesis reaction Methods 0.000 description 9
- 238000002965 ELISA Methods 0.000 description 8
- 238000011161 development Methods 0.000 description 8
- 230000018109 developmental process Effects 0.000 description 8
- 239000003112 inhibitor Substances 0.000 description 8
- 230000009467 reduction Effects 0.000 description 8
- 230000004044 response Effects 0.000 description 8
- 230000011664 signaling Effects 0.000 description 8
- 210000000987 immune system Anatomy 0.000 description 7
- 238000003068 pathway analysis Methods 0.000 description 7
- UHOVQNZJYSORNB-UHFFFAOYSA-N Benzene Chemical compound C1=CC=CC=C1 UHOVQNZJYSORNB-UHFFFAOYSA-N 0.000 description 6
- CURLTUGMZLYLDI-UHFFFAOYSA-N Carbon dioxide Chemical compound O=C=O CURLTUGMZLYLDI-UHFFFAOYSA-N 0.000 description 6
- 208000001528 Coronaviridae Infections Diseases 0.000 description 6
- 230000006378 damage Effects 0.000 description 6
- 231100000673 dose–response relationship Toxicity 0.000 description 6
- 238000002474 experimental method Methods 0.000 description 6
- 230000002757 inflammatory effect Effects 0.000 description 6
- 230000007246 mechanism Effects 0.000 description 6
- 108090000623 proteins and genes Proteins 0.000 description 6
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 6
- BTANRVKWQNVYAZ-UHFFFAOYSA-N butan-2-ol Chemical compound CCC(C)O BTANRVKWQNVYAZ-UHFFFAOYSA-N 0.000 description 5
- 239000003795 chemical substances by application Substances 0.000 description 5
- 208000035475 disorder Diseases 0.000 description 5
- 230000000694 effects Effects 0.000 description 5
- 210000002865 immune cell Anatomy 0.000 description 5
- 239000000047 product Substances 0.000 description 5
- 210000002345 respiratory system Anatomy 0.000 description 5
- 238000000926 separation method Methods 0.000 description 5
- 230000009885 systemic effect Effects 0.000 description 5
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 4
- 102000004889 Interleukin-6 Human genes 0.000 description 4
- 108090001005 Interleukin-6 Proteins 0.000 description 4
- LRHPLDYGYMQRHN-UHFFFAOYSA-N N-Butanol Chemical compound CCCCO LRHPLDYGYMQRHN-UHFFFAOYSA-N 0.000 description 4
- 229940096437 Protein S Drugs 0.000 description 4
- 206010040047 Sepsis Diseases 0.000 description 4
- 101710198474 Spike protein Proteins 0.000 description 4
- 230000001154 acute effect Effects 0.000 description 4
- 239000000556 agonist Substances 0.000 description 4
- 238000004458 analytical method Methods 0.000 description 4
- 230000008777 canonical pathway Effects 0.000 description 4
- 230000008859 change Effects 0.000 description 4
- 238000009826 distribution Methods 0.000 description 4
- 230000004064 dysfunction Effects 0.000 description 4
- 239000012091 fetal bovine serum Substances 0.000 description 4
- 238000001914 filtration Methods 0.000 description 4
- 208000027866 inflammatory disease Diseases 0.000 description 4
- 239000007788 liquid Substances 0.000 description 4
- 238000004519 manufacturing process Methods 0.000 description 4
- 230000000813 microbial effect Effects 0.000 description 4
- 238000007363 ring formation reaction Methods 0.000 description 4
- 241000114864 ssRNA viruses Species 0.000 description 4
- 230000000638 stimulation Effects 0.000 description 4
- 229940124597 therapeutic agent Drugs 0.000 description 4
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 3
- HBAQYPYDRFILMT-UHFFFAOYSA-N 8-[3-(1-cyclopropylpyrazol-4-yl)-1H-pyrazolo[4,3-d]pyrimidin-5-yl]-3-methyl-3,8-diazabicyclo[3.2.1]octan-2-one Chemical class C1(CC1)N1N=CC(=C1)C1=NNC2=C1N=C(N=C2)N1C2C(N(CC1CC2)C)=O HBAQYPYDRFILMT-UHFFFAOYSA-N 0.000 description 3
- QTBSBXVTEAMEQO-UHFFFAOYSA-N Acetic acid Chemical compound CC(O)=O QTBSBXVTEAMEQO-UHFFFAOYSA-N 0.000 description 3
- WEVYAHXRMPXWCK-UHFFFAOYSA-N Acetonitrile Chemical compound CC#N WEVYAHXRMPXWCK-UHFFFAOYSA-N 0.000 description 3
- 206010016654 Fibrosis Diseases 0.000 description 3
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 3
- 102000004457 Granulocyte-Macrophage Colony-Stimulating Factor Human genes 0.000 description 3
- 108010017213 Granulocyte-Macrophage Colony-Stimulating Factor Proteins 0.000 description 3
- 206010021143 Hypoxia Diseases 0.000 description 3
- 108010065805 Interleukin-12 Proteins 0.000 description 3
- 102000013462 Interleukin-12 Human genes 0.000 description 3
- 108010000837 Janus Kinase 1 Proteins 0.000 description 3
- 102000015617 Janus Kinases Human genes 0.000 description 3
- 108010024121 Janus Kinases Proteins 0.000 description 3
- OKKJLVBELUTLKV-UHFFFAOYSA-N Methanol Chemical compound OC OKKJLVBELUTLKV-UHFFFAOYSA-N 0.000 description 3
- 208000034486 Multi-organ failure Diseases 0.000 description 3
- KWYUFKZDYYNOTN-UHFFFAOYSA-M Potassium hydroxide Chemical compound [OH-].[K+] KWYUFKZDYYNOTN-UHFFFAOYSA-M 0.000 description 3
- DNIAPMSPPWPWGF-UHFFFAOYSA-N Propylene glycol Chemical compound CC(O)CO DNIAPMSPPWPWGF-UHFFFAOYSA-N 0.000 description 3
- 239000012980 RPMI-1640 medium Substances 0.000 description 3
- HEMHJVSKTPXQMS-UHFFFAOYSA-M Sodium hydroxide Chemical compound [OH-].[Na+] HEMHJVSKTPXQMS-UHFFFAOYSA-M 0.000 description 3
- 210000001744 T-lymphocyte Anatomy 0.000 description 3
- ZMANZCXQSJIPKH-UHFFFAOYSA-N Triethylamine Chemical compound CCN(CC)CC ZMANZCXQSJIPKH-UHFFFAOYSA-N 0.000 description 3
- 102100033438 Tyrosine-protein kinase JAK1 Human genes 0.000 description 3
- 239000002253 acid Substances 0.000 description 3
- 210000004712 air sac Anatomy 0.000 description 3
- 239000003443 antiviral agent Substances 0.000 description 3
- 238000003556 assay Methods 0.000 description 3
- 239000002585 base Substances 0.000 description 3
- 230000033228 biological regulation Effects 0.000 description 3
- 229910002092 carbon dioxide Inorganic materials 0.000 description 3
- 238000002648 combination therapy Methods 0.000 description 3
- 230000034994 death Effects 0.000 description 3
- 230000007423 decrease Effects 0.000 description 3
- 230000004069 differentiation Effects 0.000 description 3
- 239000003085 diluting agent Substances 0.000 description 3
- 210000002889 endothelial cell Anatomy 0.000 description 3
- 230000004761 fibrosis Effects 0.000 description 3
- 238000009472 formulation Methods 0.000 description 3
- 230000028993 immune response Effects 0.000 description 3
- 229940121354 immunomodulator Drugs 0.000 description 3
- 230000004957 immunoregulator effect Effects 0.000 description 3
- 230000001965 increasing effect Effects 0.000 description 3
- 208000014674 injury Diseases 0.000 description 3
- 238000011835 investigation Methods 0.000 description 3
- 238000005259 measurement Methods 0.000 description 3
- 208000029744 multiple organ dysfunction syndrome Diseases 0.000 description 3
- 244000052769 pathogen Species 0.000 description 3
- 230000007170 pathology Effects 0.000 description 3
- 239000000825 pharmaceutical preparation Substances 0.000 description 3
- 108090000765 processed proteins & peptides Proteins 0.000 description 3
- 230000002035 prolonged effect Effects 0.000 description 3
- 235000018102 proteins Nutrition 0.000 description 3
- 102000004169 proteins and genes Human genes 0.000 description 3
- 239000011347 resin Substances 0.000 description 3
- 229920005989 resin Polymers 0.000 description 3
- 229920002477 rna polymer Polymers 0.000 description 3
- 239000002904 solvent Substances 0.000 description 3
- 239000004094 surface-active agent Substances 0.000 description 3
- 238000012360 testing method Methods 0.000 description 3
- 238000002560 therapeutic procedure Methods 0.000 description 3
- 230000001960 triggered effect Effects 0.000 description 3
- 230000003827 upregulation Effects 0.000 description 3
- SPEUIVXLLWOEMJ-UHFFFAOYSA-N 1,1-dimethoxyethane Chemical compound COC(C)OC SPEUIVXLLWOEMJ-UHFFFAOYSA-N 0.000 description 2
- CIWBSHSKHKDKBQ-JLAZNSOCSA-N Ascorbic acid Chemical compound OC[C@H](O)[C@H]1OC(=O)C(O)=C1O CIWBSHSKHKDKBQ-JLAZNSOCSA-N 0.000 description 2
- 206010003598 Atelectasis Diseases 0.000 description 2
- 102100021943 C-C motif chemokine 2 Human genes 0.000 description 2
- 101710155857 C-C motif chemokine 2 Proteins 0.000 description 2
- 108010008978 Chemokine CXCL10 Proteins 0.000 description 2
- 102000006579 Chemokine CXCL10 Human genes 0.000 description 2
- 108010081668 Cytochrome P-450 CYP3A Proteins 0.000 description 2
- 102000004328 Cytochrome P-450 CYP3A Human genes 0.000 description 2
- XEKOWRVHYACXOJ-UHFFFAOYSA-N Ethyl acetate Chemical compound CCOC(C)=O XEKOWRVHYACXOJ-UHFFFAOYSA-N 0.000 description 2
- 101000763579 Homo sapiens Toll-like receptor 1 Proteins 0.000 description 2
- 101000669447 Homo sapiens Toll-like receptor 4 Proteins 0.000 description 2
- 102000014158 Interleukin-12 Subunit p40 Human genes 0.000 description 2
- 108010011429 Interleukin-12 Subunit p40 Proteins 0.000 description 2
- 108010038501 Interleukin-6 Receptors Proteins 0.000 description 2
- 102100037792 Interleukin-6 receptor subunit alpha Human genes 0.000 description 2
- 150000008575 L-amino acids Chemical class 0.000 description 2
- 208000009525 Myocarditis Diseases 0.000 description 2
- IMNFDUFMRHMDMM-UHFFFAOYSA-N N-Heptane Chemical compound CCCCCCC IMNFDUFMRHMDMM-UHFFFAOYSA-N 0.000 description 2
- 206010028813 Nausea Diseases 0.000 description 2
- CTQNGGLPUBDAKN-UHFFFAOYSA-N O-Xylene Chemical compound CC1=CC=CC=C1C CTQNGGLPUBDAKN-UHFFFAOYSA-N 0.000 description 2
- ISWSIDIOOBJBQZ-UHFFFAOYSA-N Phenol Chemical compound OC1=CC=CC=C1 ISWSIDIOOBJBQZ-UHFFFAOYSA-N 0.000 description 2
- 102000010752 Plasminogen Inactivators Human genes 0.000 description 2
- 108010077971 Plasminogen Inactivators Proteins 0.000 description 2
- 208000007123 Pulmonary Atelectasis Diseases 0.000 description 2
- 206010062106 Respiratory tract infection viral Diseases 0.000 description 2
- 238000000692 Student's t-test Methods 0.000 description 2
- DKGAVHZHDRPRBM-UHFFFAOYSA-N Tert-Butanol Chemical compound CC(C)(C)O DKGAVHZHDRPRBM-UHFFFAOYSA-N 0.000 description 2
- 102100027010 Toll-like receptor 1 Human genes 0.000 description 2
- 102100039360 Toll-like receptor 4 Human genes 0.000 description 2
- 230000010632 Transcription Factor Activity Effects 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
- 238000010521 absorption reaction Methods 0.000 description 2
- 150000007513 acids Chemical class 0.000 description 2
- 230000003213 activating effect Effects 0.000 description 2
- 239000004480 active ingredient Substances 0.000 description 2
- 239000000443 aerosol Substances 0.000 description 2
- 238000001042 affinity chromatography Methods 0.000 description 2
- XXROGKLTLUQVRX-UHFFFAOYSA-N allyl alcohol Chemical compound OCC=C XXROGKLTLUQVRX-UHFFFAOYSA-N 0.000 description 2
- 238000005349 anion exchange Methods 0.000 description 2
- 230000003110 anti-inflammatory effect Effects 0.000 description 2
- 210000000612 antigen-presenting cell Anatomy 0.000 description 2
- 238000013459 approach Methods 0.000 description 2
- 230000006793 arrhythmia Effects 0.000 description 2
- 206010003119 arrhythmia Diseases 0.000 description 2
- 230000004888 barrier function Effects 0.000 description 2
- 230000008901 benefit Effects 0.000 description 2
- DKPFZGUDAPQIHT-UHFFFAOYSA-N butyl acetate Chemical compound CCCCOC(C)=O DKPFZGUDAPQIHT-UHFFFAOYSA-N 0.000 description 2
- 229910052799 carbon Inorganic materials 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
- 238000005341 cation exchange Methods 0.000 description 2
- 230000011748 cell maturation Effects 0.000 description 2
- 210000003169 central nervous system Anatomy 0.000 description 2
- 238000006243 chemical reaction Methods 0.000 description 2
- 208000020832 chronic kidney disease Diseases 0.000 description 2
- 229960001334 corticosteroids Drugs 0.000 description 2
- 239000006184 cosolvent Substances 0.000 description 2
- 230000005860 defense response to virus Effects 0.000 description 2
- 230000001419 dependent effect Effects 0.000 description 2
- 238000001514 detection method Methods 0.000 description 2
- 229940042399 direct acting antivirals protease inhibitors Drugs 0.000 description 2
- 239000003937 drug carrier Substances 0.000 description 2
- 229940126534 drug product Drugs 0.000 description 2
- 230000008482 dysregulation Effects 0.000 description 2
- 239000002158 endotoxin Substances 0.000 description 2
- 238000005516 engineering process Methods 0.000 description 2
- 238000010201 enrichment analysis Methods 0.000 description 2
- 210000002919 epithelial cell Anatomy 0.000 description 2
- 230000006870 function Effects 0.000 description 2
- 230000037417 hyperactivation Effects 0.000 description 2
- 230000007954 hypoxia Effects 0.000 description 2
- 239000012642 immune effector Substances 0.000 description 2
- 230000036039 immunity Effects 0.000 description 2
- 230000002458 infectious effect Effects 0.000 description 2
- 230000002401 inhibitory effect Effects 0.000 description 2
- 230000015788 innate immune response Effects 0.000 description 2
- 210000005007 innate immune system Anatomy 0.000 description 2
- 238000001990 intravenous administration Methods 0.000 description 2
- 239000003446 ligand Substances 0.000 description 2
- 150000002632 lipids Chemical class 0.000 description 2
- 229920006008 lipopolysaccharide Polymers 0.000 description 2
- 210000004185 liver Anatomy 0.000 description 2
- 238000007726 management method Methods 0.000 description 2
- 239000000463 material Substances 0.000 description 2
- 230000005012 migration Effects 0.000 description 2
- 238000013508 migration Methods 0.000 description 2
- 210000001616 monocyte Anatomy 0.000 description 2
- DAZSWUUAFHBCGE-KRWDZBQOSA-N n-[(2s)-3-methyl-1-oxo-1-pyrrolidin-1-ylbutan-2-yl]-3-phenylpropanamide Chemical compound N([C@@H](C(C)C)C(=O)N1CCCC1)C(=O)CCC1=CC=CC=C1 DAZSWUUAFHBCGE-KRWDZBQOSA-N 0.000 description 2
- 230000008693 nausea Effects 0.000 description 2
- 230000007935 neutral effect Effects 0.000 description 2
- 229940021182 non-steroidal anti-inflammatory drug Drugs 0.000 description 2
- 108020004707 nucleic acids Proteins 0.000 description 2
- 102000039446 nucleic acids Human genes 0.000 description 2
- 238000012898 one-sample t-test Methods 0.000 description 2
- 210000000056 organ Anatomy 0.000 description 2
- 239000003960 organic solvent Substances 0.000 description 2
- 230000036961 partial effect Effects 0.000 description 2
- AQIXEPGDORPWBJ-UHFFFAOYSA-N pentan-3-ol Chemical compound CCC(O)CC AQIXEPGDORPWBJ-UHFFFAOYSA-N 0.000 description 2
- 239000012071 phase Substances 0.000 description 2
- 239000002797 plasminogen activator inhibitor Substances 0.000 description 2
- 239000000843 powder Substances 0.000 description 2
- 102000005962 receptors Human genes 0.000 description 2
- 108020003175 receptors Proteins 0.000 description 2
- 238000000611 regression analysis Methods 0.000 description 2
- 229940124834 selective serotonin reuptake inhibitor Drugs 0.000 description 2
- 239000012896 selective serotonin reuptake inhibitor Substances 0.000 description 2
- 210000002966 serum Anatomy 0.000 description 2
- 108091069025 single-strand RNA Proteins 0.000 description 2
- 238000001542 size-exclusion chromatography Methods 0.000 description 2
- DAEPDZWVDSPTHF-UHFFFAOYSA-M sodium pyruvate Chemical compound [Na+].CC(=O)C([O-])=O DAEPDZWVDSPTHF-UHFFFAOYSA-M 0.000 description 2
- 239000007790 solid phase Substances 0.000 description 2
- 230000006641 stabilisation Effects 0.000 description 2
- 238000011105 stabilization Methods 0.000 description 2
- 238000007619 statistical method Methods 0.000 description 2
- 239000011550 stock solution Substances 0.000 description 2
- UCSJYZPVAKXKNQ-HZYVHMACSA-N streptomycin Chemical compound CN[C@H]1[C@H](O)[C@@H](O)[C@H](CO)O[C@H]1O[C@@H]1[C@](C=O)(O)[C@H](C)O[C@H]1O[C@@H]1[C@@H](NC(N)=N)[C@H](O)[C@@H](NC(N)=N)[C@H](O)[C@H]1O UCSJYZPVAKXKNQ-HZYVHMACSA-N 0.000 description 2
- 229960005322 streptomycin Drugs 0.000 description 2
- 239000000126 substance Substances 0.000 description 2
- 239000000725 suspension Substances 0.000 description 2
- VZGDMQKNWNREIO-UHFFFAOYSA-N tetrachloromethane Chemical compound ClC(Cl)(Cl)Cl VZGDMQKNWNREIO-UHFFFAOYSA-N 0.000 description 2
- 230000001225 therapeutic effect Effects 0.000 description 2
- 230000000451 tissue damage Effects 0.000 description 2
- 231100000827 tissue damage Toxicity 0.000 description 2
- 239000003981 vehicle Substances 0.000 description 2
- 239000008096 xylene Substances 0.000 description 2
- PNBMXFUTFSFLNU-BXRBKJIMSA-N (2s)-2-aminobutanedioic acid;(2s)-2-aminopropanoic acid Chemical compound C[C@H](N)C(O)=O.OC(=O)[C@@H](N)CC(O)=O PNBMXFUTFSFLNU-BXRBKJIMSA-N 0.000 description 1
- 102000040650 (ribonucleotides)n+m Human genes 0.000 description 1
- DHKHKXVYLBGOIT-UHFFFAOYSA-N 1,1-Diethoxyethane Chemical compound CCOC(C)OCC DHKHKXVYLBGOIT-UHFFFAOYSA-N 0.000 description 1
- WSLDOOZREJYCGB-UHFFFAOYSA-N 1,2-Dichloroethane Chemical compound ClCCCl WSLDOOZREJYCGB-UHFFFAOYSA-N 0.000 description 1
- IIZPXYDJLKNOIY-JXPKJXOSSA-N 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphocholine Chemical compound CCCCCCCCCCCCCCCC(=O)OC[C@H](COP([O-])(=O)OCC[N+](C)(C)C)OC(=O)CCC\C=C/C\C=C/C\C=C/C\C=C/CCCCC IIZPXYDJLKNOIY-JXPKJXOSSA-N 0.000 description 1
- HVAUUPRFYPCOCA-AREMUKBSSA-N 2-O-acetyl-1-O-hexadecyl-sn-glycero-3-phosphocholine Chemical compound CCCCCCCCCCCCCCCCOC[C@@H](OC(C)=O)COP([O-])(=O)OCC[N+](C)(C)C HVAUUPRFYPCOCA-AREMUKBSSA-N 0.000 description 1
- 229940125673 3C-like protease inhibitor Drugs 0.000 description 1
- 102100032814 ATP-dependent zinc metalloprotease YME1L1 Human genes 0.000 description 1
- 208000004998 Abdominal Pain Diseases 0.000 description 1
- 240000005020 Acaciella glauca Species 0.000 description 1
- RZVAJINKPMORJF-UHFFFAOYSA-N Acetaminophen Chemical compound CC(=O)NC1=CC=C(O)C=C1 RZVAJINKPMORJF-UHFFFAOYSA-N 0.000 description 1
- 102000007469 Actins Human genes 0.000 description 1
- 108010085238 Actins Proteins 0.000 description 1
- 208000010470 Ageusia Diseases 0.000 description 1
- 201000004384 Alopecia Diseases 0.000 description 1
- 206010002653 Anosmia Diseases 0.000 description 1
- 206010003591 Ataxia Diseases 0.000 description 1
- 208000023275 Autoimmune disease Diseases 0.000 description 1
- 241000894006 Bacteria Species 0.000 description 1
- 208000035143 Bacterial infection Diseases 0.000 description 1
- BVKZGUZCCUSVTD-UHFFFAOYSA-M Bicarbonate Chemical compound OC([O-])=O BVKZGUZCCUSVTD-UHFFFAOYSA-M 0.000 description 1
- 102100032367 C-C motif chemokine 5 Human genes 0.000 description 1
- 102100032366 C-C motif chemokine 7 Human genes 0.000 description 1
- 101710155834 C-C motif chemokine 7 Proteins 0.000 description 1
- 101710098275 C-X-C motif chemokine 10 Proteins 0.000 description 1
- 102100036170 C-X-C motif chemokine 9 Human genes 0.000 description 1
- 108010071965 CD24 Antigen Proteins 0.000 description 1
- 102000007645 CD24 Antigen Human genes 0.000 description 1
- 102000007590 Calpain Human genes 0.000 description 1
- 108010032088 Calpain Proteins 0.000 description 1
- BVKZGUZCCUSVTD-UHFFFAOYSA-L Carbonate Chemical compound [O-]C([O-])=O BVKZGUZCCUSVTD-UHFFFAOYSA-L 0.000 description 1
- 208000031229 Cardiomyopathies Diseases 0.000 description 1
- 108010055166 Chemokine CCL5 Proteins 0.000 description 1
- 238000000665 Cohn process Methods 0.000 description 1
- 208000027932 Collagen disease Diseases 0.000 description 1
- 206010010904 Convulsion Diseases 0.000 description 1
- 241000494545 Cordyline virus 2 Species 0.000 description 1
- 208000028399 Critical Illness Diseases 0.000 description 1
- XDTMQSROBMDMFD-UHFFFAOYSA-N Cyclohexane Chemical compound C1CCCCC1 XDTMQSROBMDMFD-UHFFFAOYSA-N 0.000 description 1
- 108010037462 Cyclooxygenase 2 Proteins 0.000 description 1
- 241000701022 Cytomegalovirus Species 0.000 description 1
- 150000008574 D-amino acids Chemical class 0.000 description 1
- 108020004414 DNA Proteins 0.000 description 1
- 241000725619 Dengue virus Species 0.000 description 1
- 102000016911 Deoxyribonucleases Human genes 0.000 description 1
- 108010053770 Deoxyribonucleases Proteins 0.000 description 1
- 241000712471 Dhori virus Species 0.000 description 1
- 206010012735 Diarrhoea Diseases 0.000 description 1
- 206010060902 Diffuse alveolar damage Diseases 0.000 description 1
- 206010061818 Disease progression Diseases 0.000 description 1
- 206010059866 Drug resistance Diseases 0.000 description 1
- LVGKNOAMLMIIKO-UHFFFAOYSA-N Elaidinsaeure-aethylester Natural products CCCCCCCCC=CCCCCCCCC(=O)OCC LVGKNOAMLMIIKO-UHFFFAOYSA-N 0.000 description 1
- PIICEJLVQHRZGT-UHFFFAOYSA-N Ethylenediamine Chemical compound NCCN PIICEJLVQHRZGT-UHFFFAOYSA-N 0.000 description 1
- 208000010201 Exanthema Diseases 0.000 description 1
- 208000009386 Experimental Arthritis Diseases 0.000 description 1
- 101150021185 FGF gene Proteins 0.000 description 1
- 108010040721 Flagellin Proteins 0.000 description 1
- 206010016803 Fluid overload Diseases 0.000 description 1
- 108010010803 Gelatin Proteins 0.000 description 1
- 206010071602 Genetic polymorphism Diseases 0.000 description 1
- WHUUTDBJXJRKMK-UHFFFAOYSA-N Glutamic acid Natural products OC(=O)C(N)CCC(O)=O WHUUTDBJXJRKMK-UHFFFAOYSA-N 0.000 description 1
- 108010017080 Granulocyte Colony-Stimulating Factor Proteins 0.000 description 1
- 102000004269 Granulocyte Colony-Stimulating Factor Human genes 0.000 description 1
- 206010019233 Headaches Diseases 0.000 description 1
- 208000010496 Heart Arrest Diseases 0.000 description 1
- 206010019280 Heart failures Diseases 0.000 description 1
- 208000032843 Hemorrhage Diseases 0.000 description 1
- 208000032672 Histiocytosis haematophagic Diseases 0.000 description 1
- 101000947172 Homo sapiens C-X-C motif chemokine 9 Proteins 0.000 description 1
- 101000831567 Homo sapiens Toll-like receptor 2 Proteins 0.000 description 1
- 101000669406 Homo sapiens Toll-like receptor 6 Proteins 0.000 description 1
- 241001207270 Human enterovirus Species 0.000 description 1
- VEXZGXHMUGYJMC-UHFFFAOYSA-N Hydrochloric acid Chemical class Cl VEXZGXHMUGYJMC-UHFFFAOYSA-N 0.000 description 1
- 208000028958 Hyperferritinemia Diseases 0.000 description 1
- HEFNNWSXXWATRW-UHFFFAOYSA-N Ibuprofen Chemical compound CC(C)CC1=CC=C(C(C)C(O)=O)C=C1 HEFNNWSXXWATRW-UHFFFAOYSA-N 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
- 102000002227 Interferon Type I Human genes 0.000 description 1
- 108010014726 Interferon Type I Proteins 0.000 description 1
- 102000008070 Interferon-gamma Human genes 0.000 description 1
- 108010074328 Interferon-gamma Proteins 0.000 description 1
- 108010002586 Interleukin-7 Proteins 0.000 description 1
- 108090001007 Interleukin-8 Proteins 0.000 description 1
- 108010002335 Interleukin-9 Proteins 0.000 description 1
- 108010019437 Janus Kinase 2 Proteins 0.000 description 1
- 108010019421 Janus Kinase 3 Proteins 0.000 description 1
- WHUUTDBJXJRKMK-VKHMYHEASA-N L-glutamic acid Chemical compound OC(=O)[C@@H](N)CCC(O)=O WHUUTDBJXJRKMK-VKHMYHEASA-N 0.000 description 1
- ZDXPYRJPNDTMRX-VKHMYHEASA-N L-glutamine Chemical compound OC(=O)[C@@H](N)CCC(N)=O ZDXPYRJPNDTMRX-VKHMYHEASA-N 0.000 description 1
- 229930182816 L-glutamine Natural products 0.000 description 1
- NNJVILVZKWQKPM-UHFFFAOYSA-N Lidocaine Chemical compound CCN(CC)CC(=O)NC1=C(C)C=CC=C1C NNJVILVZKWQKPM-UHFFFAOYSA-N 0.000 description 1
- 108010028921 Lipopeptides Proteins 0.000 description 1
- 208000004852 Lung Injury Diseases 0.000 description 1
- 208000019693 Lung disease Diseases 0.000 description 1
- 206010025327 Lymphopenia Diseases 0.000 description 1
- 208000004987 Macrophage activation syndrome Diseases 0.000 description 1
- 102000005741 Metalloproteases Human genes 0.000 description 1
- 108010006035 Metalloproteases Proteins 0.000 description 1
- 241001465754 Metazoa Species 0.000 description 1
- NTIZESTWPVYFNL-UHFFFAOYSA-N Methyl isobutyl ketone Chemical compound CC(C)CC(C)=O NTIZESTWPVYFNL-UHFFFAOYSA-N 0.000 description 1
- UIHCLUNTQKBZGK-UHFFFAOYSA-N Methyl isobutyl ketone Natural products CCC(C)C(C)=O UIHCLUNTQKBZGK-UHFFFAOYSA-N 0.000 description 1
- 208000010718 Multiple Organ Failure Diseases 0.000 description 1
- 241001529936 Murinae Species 0.000 description 1
- 108010057466 NF-kappa B Proteins 0.000 description 1
- 102000003945 NF-kappa B Human genes 0.000 description 1
- CMWTZPSULFXXJA-UHFFFAOYSA-N Naproxen Natural products C1=C(C(C)C(O)=O)C=CC2=CC(OC)=CC=C21 CMWTZPSULFXXJA-UHFFFAOYSA-N 0.000 description 1
- 208000001738 Nervous System Trauma Diseases 0.000 description 1
- 241000714209 Norwalk virus Species 0.000 description 1
- 206010053159 Organ failure Diseases 0.000 description 1
- 235000010678 Paulownia tomentosa Nutrition 0.000 description 1
- 229930182555 Penicillin Natural products 0.000 description 1
- JGSARLDLIJGVTE-MBNYWOFBSA-N Penicillin G Chemical compound N([C@H]1[C@H]2SC([C@@H](N2C1=O)C(O)=O)(C)C)C(=O)CC1=CC=CC=C1 JGSARLDLIJGVTE-MBNYWOFBSA-N 0.000 description 1
- 108091005804 Peptidases Proteins 0.000 description 1
- 108010003541 Platelet Activating Factor Proteins 0.000 description 1
- 239000002202 Polyethylene glycol Substances 0.000 description 1
- 101800000795 Proadrenomedullin N-20 terminal peptide Proteins 0.000 description 1
- 102000011195 Profilin Human genes 0.000 description 1
- 108050001408 Profilin Proteins 0.000 description 1
- 102100038280 Prostaglandin G/H synthase 2 Human genes 0.000 description 1
- 239000004365 Protease Substances 0.000 description 1
- KDCGOANMDULRCW-UHFFFAOYSA-N Purine Natural products N1=CNC2=NC=NC2=C1 KDCGOANMDULRCW-UHFFFAOYSA-N 0.000 description 1
- 239000012979 RPMI medium Substances 0.000 description 1
- 208000004756 Respiratory Insufficiency Diseases 0.000 description 1
- 206010027698 Respiratory signs and symptoms Diseases 0.000 description 1
- 206010057190 Respiratory tract infections Diseases 0.000 description 1
- 102000034527 Retinoid X Receptors Human genes 0.000 description 1
- 108010038912 Retinoid X Receptors Proteins 0.000 description 1
- 102100037486 Reverse transcriptase/ribonuclease H Human genes 0.000 description 1
- 208000037847 SARS-CoV-2-infection Diseases 0.000 description 1
- 206010040070 Septic Shock Diseases 0.000 description 1
- 101710181102 Signal transducer CD24 Proteins 0.000 description 1
- 241000700584 Simplexvirus Species 0.000 description 1
- UIIMBOGNXHQVGW-DEQYMQKBSA-M Sodium bicarbonate-14C Chemical compound [Na+].O[14C]([O-])=O UIIMBOGNXHQVGW-DEQYMQKBSA-M 0.000 description 1
- 108700012920 TNF Proteins 0.000 description 1
- 208000007536 Thrombosis Diseases 0.000 description 1
- 102000008235 Toll-Like Receptor 9 Human genes 0.000 description 1
- 108010060818 Toll-Like Receptor 9 Proteins 0.000 description 1
- 102100024333 Toll-like receptor 2 Human genes 0.000 description 1
- 102100039387 Toll-like receptor 6 Human genes 0.000 description 1
- 108091023040 Transcription factor Proteins 0.000 description 1
- 102000040945 Transcription factor Human genes 0.000 description 1
- 102000004357 Transferases Human genes 0.000 description 1
- 108090000992 Transferases Proteins 0.000 description 1
- 206010069363 Traumatic lung injury Diseases 0.000 description 1
- 244000153888 Tung Species 0.000 description 1
- 102100033444 Tyrosine-protein kinase JAK2 Human genes 0.000 description 1
- 102100025387 Tyrosine-protein kinase JAK3 Human genes 0.000 description 1
- 208000024780 Urticaria Diseases 0.000 description 1
- 108010019530 Vascular Endothelial Growth Factors Proteins 0.000 description 1
- 102000005789 Vascular Endothelial Growth Factors Human genes 0.000 description 1
- 206010047700 Vomiting Diseases 0.000 description 1
- 201000006449 West Nile encephalitis Diseases 0.000 description 1
- 206010057293 West Nile viral infection Diseases 0.000 description 1
- 208000003152 Yellow Fever Diseases 0.000 description 1
- 238000009825 accumulation Methods 0.000 description 1
- 230000006978 adaptation Effects 0.000 description 1
- 230000003044 adaptive effect Effects 0.000 description 1
- 230000033289 adaptive immune response Effects 0.000 description 1
- 230000004721 adaptive immunity Effects 0.000 description 1
- 239000000654 additive Substances 0.000 description 1
- 230000000996 additive effect Effects 0.000 description 1
- 239000002671 adjuvant Substances 0.000 description 1
- 230000002411 adverse Effects 0.000 description 1
- 235000019666 ageusia Nutrition 0.000 description 1
- 150000001298 alcohols Chemical class 0.000 description 1
- 210000002588 alveolar type II cell Anatomy 0.000 description 1
- 238000005267 amalgamation Methods 0.000 description 1
- 235000001014 amino acid Nutrition 0.000 description 1
- 230000003321 amplification Effects 0.000 description 1
- 230000000202 analgesic effect Effects 0.000 description 1
- 238000000540 analysis of variance Methods 0.000 description 1
- 208000022531 anorexia Diseases 0.000 description 1
- 230000008485 antagonism Effects 0.000 description 1
- 239000003242 anti bacterial agent Substances 0.000 description 1
- 229940088710 antibiotic agent Drugs 0.000 description 1
- 229940005513 antidepressants Drugs 0.000 description 1
- 229940121357 antivirals Drugs 0.000 description 1
- 238000003491 array Methods 0.000 description 1
- 229960005070 ascorbic acid Drugs 0.000 description 1
- 235000010323 ascorbic acid Nutrition 0.000 description 1
- 239000011668 ascorbic acid Substances 0.000 description 1
- 208000006673 asthma Diseases 0.000 description 1
- 210000003719 b-lymphocyte Anatomy 0.000 description 1
- 230000001580 bacterial effect Effects 0.000 description 1
- 208000022362 bacterial infectious disease Diseases 0.000 description 1
- 239000011324 bead Substances 0.000 description 1
- 230000009286 beneficial effect Effects 0.000 description 1
- WPYMKLBDIGXBTP-UHFFFAOYSA-N benzoic acid group Chemical group C(C1=CC=CC=C1)(=O)O WPYMKLBDIGXBTP-UHFFFAOYSA-N 0.000 description 1
- MSWZFWKMSRAUBD-UHFFFAOYSA-N beta-D-galactosamine Natural products NC1C(O)OC(CO)C(O)C1O MSWZFWKMSRAUBD-UHFFFAOYSA-N 0.000 description 1
- MSWZFWKMSRAUBD-QZABAPFNSA-N beta-D-glucosamine Chemical compound N[C@H]1[C@H](O)O[C@H](CO)[C@@H](O)[C@@H]1O MSWZFWKMSRAUBD-QZABAPFNSA-N 0.000 description 1
- 229960002537 betamethasone Drugs 0.000 description 1
- UREBDLICKHMUKA-DVTGEIKXSA-N betamethasone 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-DVTGEIKXSA-N 0.000 description 1
- 230000002146 bilateral effect Effects 0.000 description 1
- 230000004071 biological effect Effects 0.000 description 1
- 230000002051 biphasic effect Effects 0.000 description 1
- 230000006406 biphasic response Effects 0.000 description 1
- 208000034158 bleeding Diseases 0.000 description 1
- 230000000740 bleeding effect Effects 0.000 description 1
- 230000023555 blood coagulation Effects 0.000 description 1
- 238000009835 boiling Methods 0.000 description 1
- 206010006451 bronchitis Diseases 0.000 description 1
- 239000000872 buffer Substances 0.000 description 1
- 244000309464 bull Species 0.000 description 1
- 238000004364 calculation method Methods 0.000 description 1
- 150000001768 cations Chemical class 0.000 description 1
- 238000004113 cell culture Methods 0.000 description 1
- 239000012578 cell culture reagent Substances 0.000 description 1
- 239000006285 cell suspension Substances 0.000 description 1
- 230000001413 cellular effect Effects 0.000 description 1
- 208000026106 cerebrovascular disease Diseases 0.000 description 1
- 239000003153 chemical reaction reagent Substances 0.000 description 1
- 239000003638 chemical reducing agent Substances 0.000 description 1
- QELSEDARGPSSRG-UHFFFAOYSA-N chlorobenzene;chloroform Chemical compound ClC(Cl)Cl.ClC1=CC=CC=C1 QELSEDARGPSSRG-UHFFFAOYSA-N 0.000 description 1
- 230000001684 chronic effect Effects 0.000 description 1
- 208000022831 chronic renal failure syndrome Diseases 0.000 description 1
- 208000035850 clinical syndrome Diseases 0.000 description 1
- 239000011248 coating agent Substances 0.000 description 1
- 238000000576 coating method Methods 0.000 description 1
- 238000004440 column chromatography Methods 0.000 description 1
- 238000002591 computed tomography Methods 0.000 description 1
- 238000007796 conventional method Methods 0.000 description 1
- 230000003436 cytoskeletal effect Effects 0.000 description 1
- 238000007405 data analysis Methods 0.000 description 1
- 206010061428 decreased appetite Diseases 0.000 description 1
- 230000003247 decreasing effect Effects 0.000 description 1
- 230000006735 deficit Effects 0.000 description 1
- 238000002716 delivery method Methods 0.000 description 1
- 210000004443 dendritic cell Anatomy 0.000 description 1
- 230000004041 dendritic cell maturation Effects 0.000 description 1
- 230000006866 deterioration Effects 0.000 description 1
- 238000003745 diagnosis Methods 0.000 description 1
- 238000009792 diffusion process Methods 0.000 description 1
- 210000002249 digestive system Anatomy 0.000 description 1
- UGMCXQCYOVCMTB-UHFFFAOYSA-K dihydroxy(stearato)aluminium Chemical compound CCCCCCCCCCCCCCCCCC(=O)O[Al](O)O UGMCXQCYOVCMTB-UHFFFAOYSA-K 0.000 description 1
- 230000005750 disease progression Effects 0.000 description 1
- 239000002270 dispersing agent Substances 0.000 description 1
- 239000006185 dispersion Substances 0.000 description 1
- 238000004821 distillation Methods 0.000 description 1
- 239000002552 dosage form Substances 0.000 description 1
- 239000006196 drop Substances 0.000 description 1
- 230000000857 drug effect Effects 0.000 description 1
- 239000012636 effector Substances 0.000 description 1
- 239000003995 emulsifying agent Substances 0.000 description 1
- 239000003797 essential amino acid Substances 0.000 description 1
- 235000020776 essential amino acid Nutrition 0.000 description 1
- LVGKNOAMLMIIKO-QXMHVHEDSA-N ethyl oleate Chemical compound CCCCCCCC\C=C/CCCCCCCC(=O)OCC LVGKNOAMLMIIKO-QXMHVHEDSA-N 0.000 description 1
- 229940093471 ethyl oleate Drugs 0.000 description 1
- 201000005884 exanthem Diseases 0.000 description 1
- 230000001747 exhibiting effect Effects 0.000 description 1
- 210000001808 exosome Anatomy 0.000 description 1
- 238000010195 expression analysis Methods 0.000 description 1
- 239000012530 fluid Substances 0.000 description 1
- 238000005194 fractionation Methods 0.000 description 1
- 239000012458 free base Substances 0.000 description 1
- 239000007789 gas Substances 0.000 description 1
- 230000002496 gastric effect Effects 0.000 description 1
- 229920000159 gelatin Polymers 0.000 description 1
- 239000008273 gelatin Substances 0.000 description 1
- 235000019322 gelatine Nutrition 0.000 description 1
- 235000011852 gelatine desserts Nutrition 0.000 description 1
- 238000003025 gene list enrichment analysis Methods 0.000 description 1
- 238000010199 gene set enrichment analysis Methods 0.000 description 1
- 239000004220 glutamic acid Substances 0.000 description 1
- 235000013922 glutamic acid Nutrition 0.000 description 1
- 239000003102 growth factor Substances 0.000 description 1
- 229940093915 gynecological organic acid Drugs 0.000 description 1
- 208000024963 hair loss Diseases 0.000 description 1
- 230000003676 hair loss Effects 0.000 description 1
- 231100000869 headache Toxicity 0.000 description 1
- 238000010438 heat treatment Methods 0.000 description 1
- 208000011316 hemodynamic instability Diseases 0.000 description 1
- 235000011167 hydrochloric acid Nutrition 0.000 description 1
- XLYOFNOQVPJJNP-UHFFFAOYSA-M hydroxide Chemical compound [OH-] XLYOFNOQVPJJNP-UHFFFAOYSA-M 0.000 description 1
- 208000018875 hypoxemia Diseases 0.000 description 1
- 229960001680 ibuprofen Drugs 0.000 description 1
- 238000003384 imaging method Methods 0.000 description 1
- 230000002519 immonomodulatory effect Effects 0.000 description 1
- 230000001900 immune effect Effects 0.000 description 1
- 229940124622 immune-modulator drug Drugs 0.000 description 1
- 239000002955 immunomodulating agent Substances 0.000 description 1
- 239000003018 immunosuppressive agent Substances 0.000 description 1
- 229940125721 immunosuppressive agent Drugs 0.000 description 1
- 238000012405 in silico analysis Methods 0.000 description 1
- 238000000126 in silico method Methods 0.000 description 1
- 238000000099 in vitro assay Methods 0.000 description 1
- 238000005462 in vivo assay Methods 0.000 description 1
- 230000006698 induction Effects 0.000 description 1
- 230000001939 inductive effect Effects 0.000 description 1
- 230000028709 inflammatory response Effects 0.000 description 1
- 206010022000 influenza Diseases 0.000 description 1
- 239000004615 ingredient Substances 0.000 description 1
- 230000000977 initiatory effect Effects 0.000 description 1
- 238000002347 injection Methods 0.000 description 1
- 239000007924 injection Substances 0.000 description 1
- 230000000266 injurious effect Effects 0.000 description 1
- 108091005434 innate immune receptors Proteins 0.000 description 1
- 229910052500 inorganic mineral Inorganic materials 0.000 description 1
- 230000002452 interceptive effect Effects 0.000 description 1
- 238000007918 intramuscular administration Methods 0.000 description 1
- 239000007951 isotonicity adjuster Substances 0.000 description 1
- 210000003292 kidney cell Anatomy 0.000 description 1
- 238000009533 lab test Methods 0.000 description 1
- 239000000787 lecithin Substances 0.000 description 1
- 229940067606 lecithin Drugs 0.000 description 1
- 235000010445 lecithin Nutrition 0.000 description 1
- 230000003902 lesion Effects 0.000 description 1
- 229960004194 lidocaine Drugs 0.000 description 1
- 230000000670 limiting effect Effects 0.000 description 1
- 239000012280 lithium aluminium hydride Substances 0.000 description 1
- 102000004311 liver X receptors Human genes 0.000 description 1
- 108090000865 liver X receptors Proteins 0.000 description 1
- 208000019423 liver disease Diseases 0.000 description 1
- 230000005976 liver dysfunction Effects 0.000 description 1
- 230000007774 longterm Effects 0.000 description 1
- 231100000515 lung injury Toxicity 0.000 description 1
- 231100001023 lymphopenia Toxicity 0.000 description 1
- 210000002540 macrophage Anatomy 0.000 description 1
- 238000012423 maintenance Methods 0.000 description 1
- 239000003550 marker Substances 0.000 description 1
- 229910052751 metal Inorganic materials 0.000 description 1
- 239000002184 metal Substances 0.000 description 1
- 229940043265 methyl isobutyl ketone Drugs 0.000 description 1
- 108091050864 miR-1a stem-loop Proteins 0.000 description 1
- 230000003278 mimic effect Effects 0.000 description 1
- 235000010755 mineral Nutrition 0.000 description 1
- 239000011707 mineral Substances 0.000 description 1
- 150000007522 mineralic acids Chemical class 0.000 description 1
- 238000005065 mining Methods 0.000 description 1
- 238000012544 monitoring process Methods 0.000 description 1
- 238000011294 monotherapeutic Methods 0.000 description 1
- VCWNAJUHTLWQQT-YRGDBDHISA-N mpma Chemical compound C([C@@]1(C(=O)C(C)=CC1[C@@]1(O)[C@H](C)[C@H]2OC(=O)CCCCCCCCCCCCC)OC)C(CO)=C[C@H]1C1[C@]2(OC(C)=O)C1(C)C VCWNAJUHTLWQQT-YRGDBDHISA-N 0.000 description 1
- 201000006417 multiple sclerosis Diseases 0.000 description 1
- 210000000066 myeloid cell Anatomy 0.000 description 1
- 208000037891 myocardial injury Diseases 0.000 description 1
- ACTNHJDHMQSOGL-UHFFFAOYSA-N n',n'-dibenzylethane-1,2-diamine Chemical compound C=1C=CC=CC=1CN(CCN)CC1=CC=CC=C1 ACTNHJDHMQSOGL-UHFFFAOYSA-N 0.000 description 1
- 229960002009 naproxen Drugs 0.000 description 1
- CMWTZPSULFXXJA-VIFPVBQESA-N naproxen Chemical compound C1=C([C@H](C)C(O)=O)C=CC2=CC(OC)=CC=C21 CMWTZPSULFXXJA-VIFPVBQESA-N 0.000 description 1
- 210000000822 natural killer cell Anatomy 0.000 description 1
- 238000002663 nebulization Methods 0.000 description 1
- 208000028412 nervous system injury Diseases 0.000 description 1
- 208000004296 neuralgia Diseases 0.000 description 1
- 210000000440 neutrophil Anatomy 0.000 description 1
- 231100000252 nontoxic Toxicity 0.000 description 1
- 230000003000 nontoxic effect Effects 0.000 description 1
- 238000010606 normalization Methods 0.000 description 1
- 238000003199 nucleic acid amplification method Methods 0.000 description 1
- 150000007523 nucleic acids Chemical class 0.000 description 1
- 235000008390 olive oil Nutrition 0.000 description 1
- 239000004006 olive oil Substances 0.000 description 1
- 230000009210 ongoing activation Effects 0.000 description 1
- 150000007524 organic acids Chemical class 0.000 description 1
- 235000005985 organic acids Nutrition 0.000 description 1
- 150000002892 organic cations Chemical class 0.000 description 1
- 150000002895 organic esters Chemical class 0.000 description 1
- 230000001151 other effect Effects 0.000 description 1
- 229940094443 oxytocics prostaglandins Drugs 0.000 description 1
- 229960005489 paracetamol Drugs 0.000 description 1
- 239000002245 particle Substances 0.000 description 1
- 230000001717 pathogenic effect Effects 0.000 description 1
- 230000001575 pathological effect Effects 0.000 description 1
- 230000007310 pathophysiology Effects 0.000 description 1
- 238000003909 pattern recognition Methods 0.000 description 1
- 229940049954 penicillin Drugs 0.000 description 1
- 238000002837 percent inhibition normalization Methods 0.000 description 1
- 239000000546 pharmaceutical excipient Substances 0.000 description 1
- 229940127557 pharmaceutical product Drugs 0.000 description 1
- WVDDGKGOMKODPV-ZQBYOMGUSA-N phenyl(114C)methanol Chemical compound O[14CH2]C1=CC=CC=C1 WVDDGKGOMKODPV-ZQBYOMGUSA-N 0.000 description 1
- 239000008363 phosphate buffer Substances 0.000 description 1
- 230000036470 plasma concentration Effects 0.000 description 1
- 230000010287 polarization Effects 0.000 description 1
- 229920001223 polyethylene glycol Polymers 0.000 description 1
- 229920005862 polyol Polymers 0.000 description 1
- 150000003077 polyols Chemical class 0.000 description 1
- 238000002360 preparation method Methods 0.000 description 1
- 102000004196 processed proteins & peptides Human genes 0.000 description 1
- 238000004393 prognosis Methods 0.000 description 1
- 208000037821 progressive disease Diseases 0.000 description 1
- 239000003380 propellant Substances 0.000 description 1
- 150000003180 prostaglandins Chemical class 0.000 description 1
- 125000006239 protecting group Chemical group 0.000 description 1
- 201000001474 proteinuria Diseases 0.000 description 1
- 206010037844 rash Diseases 0.000 description 1
- 230000036632 reaction speed Effects 0.000 description 1
- 235000003499 redwood Nutrition 0.000 description 1
- 238000010992 reflux Methods 0.000 description 1
- 230000008085 renal dysfunction Effects 0.000 description 1
- 230000010076 replication Effects 0.000 description 1
- 238000011160 research Methods 0.000 description 1
- 201000004193 respiratory failure Diseases 0.000 description 1
- 230000004202 respiratory function Effects 0.000 description 1
- 230000036303 septic shock Effects 0.000 description 1
- 239000012679 serum free medium Substances 0.000 description 1
- 238000009097 single-agent therapy Methods 0.000 description 1
- 229910052708 sodium Inorganic materials 0.000 description 1
- 239000011734 sodium Substances 0.000 description 1
- 229940054269 sodium pyruvate Drugs 0.000 description 1
- 241000894007 species Species 0.000 description 1
- 239000007921 spray Substances 0.000 description 1
- 238000007920 subcutaneous administration Methods 0.000 description 1
- 235000000346 sugar Nutrition 0.000 description 1
- 150000008163 sugars Chemical class 0.000 description 1
- 239000006228 supernatant Substances 0.000 description 1
- 230000003319 supportive effect Effects 0.000 description 1
- 230000002195 synergetic effect Effects 0.000 description 1
- 230000002194 synthesizing effect Effects 0.000 description 1
- 238000010189 synthetic method Methods 0.000 description 1
- 238000010257 thawing Methods 0.000 description 1
- 201000005665 thrombophilia Diseases 0.000 description 1
- 125000003944 tolyl group Chemical group 0.000 description 1
- 238000013518 transcription Methods 0.000 description 1
- 230000035897 transcription Effects 0.000 description 1
- 230000009466 transformation Effects 0.000 description 1
- 230000008733 trauma Effects 0.000 description 1
- 238000011277 treatment modality Methods 0.000 description 1
- 201000008827 tuberculosis Diseases 0.000 description 1
- 230000014567 type I interferon production Effects 0.000 description 1
- 230000002568 urticarial effect Effects 0.000 description 1
- 230000002792 vascular Effects 0.000 description 1
- 230000008728 vascular permeability Effects 0.000 description 1
- 235000015112 vegetable and seed oil Nutrition 0.000 description 1
- 239000008158 vegetable oil Substances 0.000 description 1
- 238000009423 ventilation Methods 0.000 description 1
- 230000008299 viral mechanism Effects 0.000 description 1
- 230000008673 vomiting Effects 0.000 description 1
- 239000000080 wetting agent Substances 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/12—Antivirals
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/495—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/495—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
- A61K31/505—Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
- A61K31/519—Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/56—Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
- A61K31/57—Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone
- A61K31/573—Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone substituted in position 21, e.g. cortisone, dexamethasone, prednisone or aldosterone
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/70—Carbohydrates; Sugars; Derivatives thereof
- A61K31/7042—Compounds having saccharide radicals and heterocyclic rings
- A61K31/7052—Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
- A61K31/706—Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/17—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- A61K38/38—Albumins
- A61K38/385—Serum albumin
-
- 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
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/12—Antivirals
- A61P31/14—Antivirals for RNA viruses
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D241/00—Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings
- C07D241/02—Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings
- C07D241/06—Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings having one or two double bonds between ring members or between ring members and non-ring members
- C07D241/08—Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings having one or two double bonds between ring members or between ring members and non-ring members with oxygen atoms directly attached to ring carbon atoms
Definitions
- the present disclosure relates to methods of treating TLR7/8-mediated disease, including viral respiratory disease.
- the methods comprise administering an effective amount of a pharmaceutical composition prepared by removing albumin from a solution of a human serum albumin composition and/or comprising a diketopiperazine with amino acid side chains of aspartic acid and alanine (DA-DKP), such as a low molecular weight fraction of human serum albumin.
- a pharmaceutical composition prepared by removing albumin from a solution of a human serum albumin composition and/or comprising a diketopiperazine with amino acid side chains of aspartic acid and alanine (DA-DKP), such as a low molecular weight fraction of human serum albumin.
- DAKP diketopiperazine with amino acid side chains of aspartic acid and alanine
- TLRs Toll-like receptors
- Treatments that modulate and/or interrupt TLR signaling may prove beneficial for the treatment of diseases, for example when immune cells are activated by antiviral mechanisms that may help drive hyperinflammation in patients with viral infections, especially respiratory viruses.
- immune cells are activated by antiviral mechanisms that may help drive hyperinflammation in patients with viral infections, especially respiratory viruses.
- SARS-CoV- 2 severe acute respiratory syndrome coronavirus 2
- SARS-CoV-2 replication is primarily localized to the upper respiratory tract, with limited innate immune response and low viral burden.
- underlying factors such as genetic polymorphisms, autoantibody development, and intrinsic viral mechanisms have been identified that serve to suppress or delay type I interferon production or activity during the course of infection.
- the resulting maladaptation in antiviral immunity helps drive progression into more severe stages marked by migration of the virus into the lower respiratory tract, elevated viral loads, and dramatic loss of type II pneumocytes.
- TLRs innate immune effector mechanisms
- pro-inflammatory cytokines like CXCL10 become predictive of the clinical course, and approximately 20% of these patients go on to develop acute respiratory distress syndrome (ARDS).
- ARDS acute respiratory distress syndrome
- these critical stages are characterized by increased plasma levels of pro- inflammatory cytokines and chemokines, such as IFNy, CXCL10, IL-Ib, and TNFa, indicative of “cytokine storm” development that can eventually lead to multiorgan failure and death.
- ARDS acute respiratory distress syndrome
- pro- inflammatory cytokines and chemokines such as IFNy, CXCL10, IL-Ib, and TNFa
- Diketopiperazines have been reported to exhibit a variety of biological activities. See, e.g, U.S. Patent Nos. 4,289,759 (immunoregulatory agents), 4,331,595 (immunoregulatory agents), 4,940,709 (PAF antagonists), 5,700,804 (inhibitors of plasminogen activator inhibitor), 5,750,530 (inhibitors of plasminogen activator inhibitor) and 5,990,112 (inhibitors of metalloproteases); PCT publication nos. WO 97/36888 (inhibitors of farnesyl-protein transferase) and WO 99/40931 (treatment of central nervous system injury); EP Patent No.
- compositions and methods useful to treat diseases mediated by TLR7 and/or TLR8, which recognize single-stranded (ssRNA) RNA viruses are disclosed.
- the disclosed compositions and methods are useful to treat symptoms caused by infection with ssRNA viruses and viruses recognized by TLR7 and/or TLR8.
- the compositions and methods disclosed herein are useful to treat inflammation caused by infection with a virus, for example a respiratory virus and/or a ssRNA respiratory virus.
- the compositions and methods disclosed herein are also useful to treat other diseases mediated by TLR7 and/or TLR8, such as lupus and/or lupus nephritis.
- compositions and methods useful to treat or prevent one or more symptoms of a viral infection using a biphasic or multi-modal treatment method including treating inflammation during a hyperinflammatory stage of a TLR7- and/or TLR8- mediated disease.
- treatment of a patient occurs by administration of a first drug, during the acute phase of the disease, followed by administration of a pharmaceutical composition comprising DA-DKP before, at or after the onset of a hyperinflammatory stage of the disease.
- the first drug and the pharmaceutical composition are co-administered to the patient.
- the first drug is an antiviral, an immune- modifying drug, an anti-depressant, a corticosteroid, or combinations thereof, among other things.
- a method of treating one or more symptoms of a viral infection in a patient comprises administering a first drug to the patient prior to onset of a hyperinflammatory stage of the infection and administering a pharmaceutical composition comprising DA-DKP to the patient before, at, or after the onset of the hyperinflammatory stage.
- a method of treating inflammation during a hyperinflammatory stage of a TLR7- and/or TLR8-mediated disease in a patient comprises administering a first drug to the patient prior to onset of a hyperinflammatory stage of the disease and administering a pharmaceutical composition comprising DA-DKP to the patient before, at, or after the onset of the hyperinflammatory stage.
- a method of treating or preventing one or more symptoms of a TLR7- and/or TLR8-mediated disease in a patient, by administering a pharmaceutical composition comprising DA-DKP is disclosed.
- a method of treating or preventing inflammation associated with a TLR7- and/or TLR8-mediated disease in a patient, by administering a pharmaceutical composition comprising DA-DKP is disclosed.
- FIGS. 1A and IB show data demonstrating that AMPION ® inhibits CXCL10 release from THP-1 cells.
- Data are presented as mean pg/ml CXCL10 ⁇ STD of three technical replicates 24 hours (FIG. 1 A) or 72 hours (FIG. IB) post stimulation.
- * p-value ⁇ 0.05 vs activated saline control by student t-test.
- FIGS. 2A and 2B show data demonstrating that AMPION ® inhibits CXCL10 release from THP-1 cells.
- Data are presented as mean pg/ml CXCL10 ⁇ STD of three technical replicates 24 hours (FIG. 2A) or 72 hours (FIG. 2B) post stimulation.
- * p-value ⁇ 0.05 vs activated saline control by student t-test.
- FIGS. 3A, 3B, and 3C show data demonstrating that AMPION ® inhibits CXCL10 release in THP-1 cells and in Peripheral Blood Mononuclear Cells (PBMC). Box plots for 24-hour 5 pg/ml CL075-induced CXCL10 release and AMPION ® percent inhibitions for PMA-differentiated THP-1 cells and PBMC are shown. Data are presented as CL075- induced CXCL10 pg/ml release for both saline- and AMPION ® -treatment groups from PMA-differentiated THP-1 (FIG. 3 A) or PBMC (FIG.
- PBMC Peripheral Blood Mononuclear Cells
- FIG. 4 shows data from a relative potency assay of AMPION ® inhibition of CXCL10.
- the data show dose-dependent reduction in CL075-induced CXCL10 release from PMA-differentiated THP-1 by AMPION ® .
- FIG. 5 shows canonical pathway analysis that demonstrates AMPION ® directional regulation of cytokines and chemokines are predicted to inhibit pathways associated with increased inflammation.
- Log2 fold-changes and p-values of cytokines and chemokines from the 48-plex cytokine array comparing saline-treated versus AMPION ® -treated PMA- differentiated THP-1 cells activated with 5 pg/ml CL075 for 24h were uploaded into IPA for canonical pathway analysis.
- LMWF refers to a Low Molecular Weight Fraction of human serum albumin (HSA) that is a composition prepared by separation of high molecular weight components from HSA.
- HSA human serum albumin
- LMWF can be prepared by filtration of commercially available HSA solutions wherein molecular weight components of more than 3 kDa, more than 5 kDa, more than 10 kDa, more than 20 kDa, more than 30 kDa, more than 40 kDa, or more than 50 kDa are separated from the HSA solution.
- LMWF refers to a composition prepared by separation of high molecular weight components from HSA by other techniques, including but not limited to ultrafiltration, column chromatography including size exclusion chromatography, affinity chromatography, anion exchange, cation exchange, sucrose gradient centrifugation, salt precipitation, and/or sonication.
- LMWF also refers to a composition that includes components of HSA having a molecular weight less than 50 kDa, less than 40 kDa, less than 30 kDa, less than 20 kDa, less than 10 kDa, less than 5000 Da, less than 4000 Da, or less than 3000 Da (corresponding to 50,000 g/mol, 40,000 g/ml, 30,000 g/mol, 20,00 g/mol, 10,000 g/mol, 5,000 g/mol, 4,000 g/mol or 3,000 g/mol, respectively).
- the terms “patient” and “subject” are interchangeable and generally refer to an animal or a human to which a composition disclosed herein is administered or is to be administered.
- prodrug refers to derivatives of a pharmacologically active drug molecule which undergo transformation within the body to produce the pharmacologically active drug, also referred to sometimes as the “parent” drug. Prodrugs are frequently, although not necessarily, pharmacologically inactive until converted to the parent drug.
- a “therapeutically effective amount,” “effective amount,” or the like means the amount of a compound or composition that, when administered to a patient for treating a disease, is sufficient to effect such treatment for the disease.
- the “effective amount” will vary depending on the compound, the disease and its severity and the age, weight, etc., of the patient to be treated.
- the terms “treat,” “treatment,” “treating,” and derivatives thereof mean to reduce (wholly or partially) the symptoms, duration or severity of a disease. In some embodiments, such terms relate to ameliorating a disease or disorder (/. ., arresting or reducing the development of the disease or at least one of the clinical symptoms thereof).
- such terms refer to ameliorating at least one physical parameter, which may or may not be discernible by the patient.
- such terms refer to inhibiting the disease or disorder, either physically, (e.g., stabilization of a discernible symptom), physiologically, (e.g., stabilization of a physical parameter), or both.
- “treating” or “treatment” refers to delaying the onset of the disease or disorder. In accordance with the present state of the art, treat typically does not mean to cure.
- the present disclosure generally relates to methods of treating a viral respiratory disease, for example a TLR7/8-associated disease such as COVID-19, and/or one or more symptoms thereof.
- a viral respiratory disease for example a TLR7/8-associated disease such as COVID-19
- the methods of treatment relate to the administration of the disclosed compositions as mono-therapeutic agents to treat viral respiratory diseases.
- the methods of treatment relate to the administration of the disclosed compositions in combination with one or more additional therapeutics for the treatment of a viral respiratory disease.
- the methods comprise administering an effective amount of a pharmaceutical composition comprising aspartyl-alanyl diketopiperazine (DA- DKP) to a subject having a need thereof.
- DA- DKP aspartyl-alanyl diketopiperazine
- the pharmaceutical composition can be prepared by removing albumin from a solution of a human serum albumin composition.
- the pharmaceutical composition can also include N-acetyl-tryptophan (NAT), caprylic acid, caprylate or combinations thereof.
- the pharmaceutical composition can comprise AMPION ® (defined herein).
- the methods comprise administering an effective amount of a pharmaceutical composition comprising DA-DKP before, during, or after the initiation of a hyperinflammatory stage of viral disease to a subject having a need thereof.
- the pharmaceutical composition can be prepared as noted above, by removing albumin from a solution of a human serum albumin composition.
- the pharmaceutical composition can also include N-acetyl-tryptophan (NAT), caprylic acid, caprylate or combinations thereof.
- the pharmaceutical composition can comprise AMPION ® (defined herein).
- the present disclosure generally relates to methods of treating one or more respiratory viral diseases by administering an effective amount of a pharmaceutical composition comprising DA-DKP before a hyperinflammatory stage of respiratory viral disease to a subject having a need thereof.
- the pharmaceutical composition can be prepared as noted above, by removing albumin from a solution of a human serum albumin composition.
- the pharmaceutical composition can also include N-acetyl-tryptophan (NAT), caprylic acid, caprylate or combinations thereof.
- the pharmaceutical composition can comprise AMPION ® (defined herein).
- the present disclosure relates to methods of treating COVID-19 by administering an effective amount of a pharmaceutical composition comprising DA- DKP before a hyperinflammatory stage of COVID-19 to a subject having a need thereof.
- the pharmaceutical composition can be prepared as noted above, by removing albumin from a solution of a human serum albumin composition.
- the pharmaceutical composition can also include N-acetyl-tryptophan (NAT), caprylic acid, caprylate or combinations thereof.
- the pharmaceutical composition can comprise AMPION ® (defined herein).
- the present disclosure relates to methods of treating viral diseases by administering an effective amount of a pharmaceutical composition comprising DA-DKP during a hyperinflammatory stage of viral disease to a subject having a need thereof.
- the pharmaceutical composition can be prepared as noted above, by removing albumin from a solution of a human serum albumin composition.
- the pharmaceutical composition can also include N-acetyl-tryptophan (NAT), caprylic acid, caprylate or combinations thereof.
- the pharmaceutical composition can comprise AMPION ® (defined herein).
- the present disclosure relates to methods of treating COVID-19 by administering an effective amount of a pharmaceutical composition comprising DA- DKP during a hyperinflammatory stage of COVID-19 to a subject having a need thereof.
- the pharmaceutical composition can be prepared as noted above, by removing albumin from a solution of a human serum albumin composition.
- the pharmaceutical composition can also include N-acetyl-tryptophan (NAT), caprylic acid, caprylate or combinations thereof.
- the pharmaceutical composition can comprise AMPION ® (defined herein).
- the present disclosure relates to methods of treating viral diseases by administering an effective amount of a pharmaceutical composition comprising DA-DKP to a subject after a hyperinflammatory stage of viral disease has begun.
- the pharmaceutical composition can be prepared as noted above, by removing albumin from a solution of a human serum albumin composition.
- the pharmaceutical composition can also include N-acetyl-tryptophan (NAT), caprylic acid, caprylate or combinations thereof.
- the pharmaceutical composition can comprise AMPION ® (defined herein).
- the present disclosure relates to methods of treating COVID-19 by administering an effective amount of a pharmaceutical composition comprising DA- DKP to a subject after a hyperinflammatory stage of COVID-19 has begun.
- the pharmaceutical composition can be prepared as noted above, by removing albumin from a solution of a human serum albumin composition.
- the pharmaceutical composition can also include N-acetyl-tryptophan (NAT), caprylic acid, caprylate or combinations thereof.
- the pharmaceutical composition can comprise AMPION ® (defined herein).
- DA-DKP has multiple anti-inflammatory and immune modulating effects including inhibition of multiple pro-inflammatory cytokines, chemokines and signaling molecules at the transcription level, inhibition of the migration and adhesion of T-cells and monocytes, activity at the G-coupled protein receptor level, activity on actin-dependent cytoskeletal events, and reduction in vascular permeability and inhibition of inflammation induced by platelet activating factor, among other effects.
- DA-DKP can be used to treat lung inflammatory diseases, inflammation related to viral respiratory diseases, and/or to prevent lung inflammation and lung inflammatory diseases related to viral respiratory diseases.
- DA- DKP can also be used to prevent, reduce in severity and/or duration, and/or otherwise treat cytokine storm or cytokine release syndrome. Cytokine storm or cytokine release syndrome is a systemic inflammatory syndrome characterized by elevated levels of circulating cytokines and immune cell hyperactivation. Innate immune system and Toll-like Receptors
- TLR Toll-like receptors
- TLRs Toll-like receptors
- the human genome includes 10 known TLRs.
- the ligands for these receptors are highly conserved microbial molecules such as lipopolysaccharides (LPS) (recognized by TLR4), lipopeptides (TLR2 in combination with TLR1 or TLR6), flagellin (TLR5), single stranded RNA (TLR7 and TLR8), double stranded RNA (TLR3), CpG motif- containing DNA (recognized by TLR9), and profilin present on uropathogenic bacteria (TLR 11).
- LPS lipopolysaccharides
- TLR2 lipopeptides
- TLR5 flagellin
- TLR7 and TLR8 single stranded RNA
- TLR3 double stranded RNA
- CpG motif- containing DNA profilin present on uropathogenic bacteria
- TLR1, -2, -4, -5, and -6 respond to extracellular stimuli
- TLR3, -7, -8 and -9 respond to intracytoplasmic pathogen-associated molecular patterns (PAMPs, conserved microbial small molecular motifs).
- PAMPs pathogen-associated molecular patterns
- TLRs by their cognate ligands leads to activation of innate immune effector mechanisms, including the production of pro- inflammatory cytokines, and up-regulation of MHC molecules and co-stimulatory signals in antigen-presenting cells as well as activating natural killer (NK) cells.
- NK natural killer
- Toll-like receptors -7/-8 are innate immune receptors present in the endosomal compartment that are activated by single-stranded RNA (ssRNA) molecules of viral as well as nonviral origin, inducing the production of inflammatory cytokines necessary for the development of adaptive immunity.
- ssRNA single-stranded RNA
- TLR7 is an endosomal sensor that can discriminate non-self, single-stranded ribonucleic acid sequences to trigger an anti-viral response. TLR 7 serves to detect non-self, single stranded ribonucleic acid, and trigger anti-viral immunity.
- dysregulation of TLR7 signaling has been linked to the pathology of inflammatory diseases that present with cytokine storms and acute kidney injury such as sepsis and lupus.
- TLR7 is linked to plasma cytokine storm development, acute kidney injury, and mortality.
- Cytokine storm is a systemic inflammatory syndrome characterized by high levels of circulating cytokines and hyperactivation of the immune system. This can lead to multiple organ failure and even death.
- a cytokine storm, or cytokine storm syndrome can be caused by many infectious and non-infectious etiologies.
- cytokine storm syndrome can be caused by bacterial and/or viral infection, such as infection by the coronavirus SARS- CoV-2, which causes the disease COVID-19.
- Cytokine storm symptoms include fever, fatigue, nausea, systemic inflammation, hyperferritinemia, hemodynamic instability, organ failure, and death.
- Cytokine storm can damage the heart, kidneys, lungs, liver, central nervous system, and other organs, for example by causing cardiac arrest, myocarditis, myocardial injury, cardiomyopathy, arrhythmia, acute kidney injury, proteinuria, and acute respiratory distress syndrome. Furthermore, infection, for example by SARS-CoV-2, can lead to sepsis, which is associated with cytokine storm. Severe COVID-19 patients exhibit cytokine storms associated with acute kidney injury and renal dysfunction.
- Cytokine storm can be characterized by elevated levels of cytokines including IL-Ib, IL-7, IL-8, IL-9, IL- 10, FGF, G-CSF, GM-CSF, IFN-g, IP-10, MCP-1, MPMA, MIP1-B, PDGF, TNF-a, and VEGF.
- cytokines including IL-Ib, IL-7, IL-8, IL-9, IL- 10, FGF, G-CSF, GM-CSF, IFN-g, IP-10, MCP-1, MPMA, MIP1-B, PDGF, TNF-a, and VEGF.
- TLR7 is also linked to the induction of serum autoantibody products that accelerate glomerulonephritis, exacerbate macrophage activation syndrome, and result in rapid mortality in murine lupus models.
- Lupus nephritis is linked to TLR7.
- Lupus nephritis is an autoimmune disease caused in part by the recognition of kidney cells by autoantibodies.
- compositions provided by the present disclosure are useful for the treatment of lupus, including lupus nephritis. Such embodiments are exemplified by methods that can comprise administering an effective amount of a pharmaceutical composition comprising aspartyl-alanyl diketopiperazine (DA-DKP) to a subject having a need thereof.
- the pharmaceutical composition can be prepared by removing albumin from a solution of a human serum albumin composition.
- the pharmaceutical composition can also include N-acetyl-tryptophan (NAT), caprylic acid, caprylate or combinations thereof.
- the pharmaceutical composition can comprise AMPION ® (defined herein).
- ssRNA viruses include Coronaviridae (e.g ., SARS-CoV, MERS, SARS-CoV-2, and toroviruses), Hepeviridae (e.g., Hepatitis E), Caliciviridae (e.g., Norovirus, formerly Norwalk virus, and Sapporo virus), Togaviridae (e.g ., Alphavirus viral diseases (e.g., Chikungunya, Eastern equine encephalomyelitis virus, Getah virus, Mayaro virus, Mucambo virus, O’nyong’nyong virus, Ross river virus, Barmah forest virus, Sagiyama virus, Semliki forest virus, Sindbis virus, Tonate virus, Venezuelan equine encephalomyelitis virus, Western equine encephal
- Coronaviridae e.g ., SARS-CoV, MERS, SARS-CoV-2, and toroviruses
- TLR7 and/or TLR8 Diseases mediated by TLR7 and/or TLR8 are diseases caused by infection with ssRNA viruses recognized by TLR7 and/or TLR8, such as inflammation caused by a ssRNA virus, as well as lupus and lupus nephritis.
- a complication of disease caused by viral infection is overactive immune response, leading to excessive inflammation. This can lead to development of a hyperinflammatory stage of infection that is characterized by cytokine storm, acute lung injury (ALI), and/or acute respiratory distress syndrome (ARDS).
- cytokine storm acute lung injury
- ARDS acute respiratory distress syndrome
- Many viruses can cause excessive inflammation, cytokine storm, ALI, and/or ARDS, including respiratory viruses, Herpes simplex viruses, and cytomegaloviruses. Certain respiratory viruses are particularly likely to cause excessive inflammation and even ARDS, such as viruses that cause community- acquired pneumonia and/or pandemics.
- Respiratory viruses that are especially likely to cause ARDS include influenza viruses, coronaviruses, rhinoviruses, respiratory syncytial viruses, parainfluenzaviruses, human metapneumoviruses, and adenoviruses. These include influenza A viruses (including H1N1 and H3N2 strains), influenza B viruses, Severe Acute Respiratory Distress Syndrome coronavirus (SARS-CoV), Middle East Respiratory Syndrome coronavirus (MERS-CoV), Severe Acute Respiratory Distress Syndrome coronavirus 2 (SARS-CoV-2), and human adenovirus B21 infections (HAdV-B21). Other emergent viruses, especially those that cause community-acquired pneumonia and/or pandemics, are likely to also cause ARDS. In various aspects, compositions provided by the present disclosure are useful to treat ARDS caused by any virus.
- a viral respiratory disease is an illness caused by a virus that affects the respiratory tract.
- viral respiratory diseases can include Severe Acute Respiratory Distress Syndrome (SARS), Middle East Respiratory Syndrome (MERS), COVID-19, and viral infections associated with asthma, pneumonia, bronchitis and/or tuberculosis.
- Viruses that can cause one or more viral respiratory diseases include coronaviruses, influenza viruses, respiratory syncytial virus (RSV), parainfluenza viruses, and respiratory adenoviruses.
- Coronaviruses include SARS-Coronavirus-2 (SARS-CoV-2), SARS-associated coronavirus (SARS-CoV), and Middle East Respiratory Syndrome Coronavirus (MERS- CoV).
- Coronavirus infections and other viral infections can cause acute respiratory distress syndrome (ARDS), acute lung injury (ALI), interstitial lung disease, pulmonary fibrosis, pneumonia, and reactive airway disease syndrome.
- Coronavirus infections and other viral infections can cause inflammation in tissues such as lung, brain, heart, kidney, blood vessel, skin, and nerve.
- Coronavirus infections and other viral infections can cause symptoms such as fatigue, shortness of breath or difficulty breathing, low exercise tolerance, low blood oxygen saturation, cough, sore throat, stuffy or runny nose, joint pain, chest pain, tightness, or discomfort, muscle pain, muscle weakness, fever, heart palpitations, difficulty thinking and/or concentrating, and depression.
- COVID-19 infection is a respiratory illness caused by the novel coronavirus SARS- COV-2 and has been classified as a pandemic with no known cure to date. COVID-19 is detected and diagnosed with a laboratory test. The primary symptoms of COVID-19 infection include mild symptoms such as fever, cough, chills, muscle pain, headache, gastrointestinal symptoms, and the loss of taste or smell. Once infected, the virus moves down a patient’s respiratory tract, where the lungs may become inflamed, making breathing difficult and sometimes requiring supplemental oxygen in the more severe cases of the disease.
- Respiratory symptoms after a COVID-19 infection include shortness of breath, cough, chest discomfort, low exercise tolerance and low oxygen saturation, all of which point to potential inflammation-related complication sequelae.
- Infiltrating or resident cells in the immune system e.g., macrophages, peripheral blood mononuclear cells, etc.
- Chronic or prolonged inflammation of the lungs may be responsible for a myriad of respiratory signs and symptoms experienced by patients after a COVID-19 infection.
- Chest x-rays and CT scans reveal disturbing patterns of perhaps extensive fibrosis and potential loss of elasticity and oxygen diffusion capacity.
- the SARS-Cov-2 virus transmits through the respiratory system and can cause a severe dysregulation of the immune response and damage in the lungs.
- the hyperinflammatory state is thought to lead to prolonged clinical complications, and treatment with immunomodulators at this later point in the disease can be more effective than anti-viral treatment.
- ARDS acute lung injury
- ARDS acute respiratory distress syndrome
- ALI acute lung injury
- ARDS acute respiratory distress syndrome
- ARDS is associated with widespread inflammation in the lungs.
- the underlying mechanism of ARDS involves diffuse injury to cells which form the barrier of the microscopic air sacs (alveoli) of the lung, surfactant dysfunction, and activation of the immune system.
- the fluid accumulation in the lungs associated with ARDS is partially explained by vascular leakage due to inflammation.
- An important aspect of ARDS, triggered by COVID-19, is an initial release of chemical signals and other inflammatory mediators secreted by lung epithelial and endothelial cells.
- COVID-19 Adaptation recommends early intervention with supplemental oxygen for COVID-19 patients with low blood oxygen saturation (Sp02) beginning with the least invasive modality possible (e.g . hand-held oxygen source) and moving to more invasive modalities (e.g. bilevel positive airway pressure [BiPAP] and/or non-invasive ventilation (NIV)) as severity increases.
- invasive modality e.g. hand-held oxygen source
- invasive modalities e.g. bilevel positive airway pressure [BiPAP] and/or non-invasive ventilation (NIV)
- Treatment during early intervention for COVID-19 patients with respiratory distress requires monitoring of respiratory function with treatment responsive to disease progression.
- the CDC recommends following the guidelines for treatment of COVID-19 patients with hypoxia in Surviving Sepsis Campaign: Guidelines on the Management of Critically Ill Adults with COVID-19.
- ARDS a rapidly progressive disease characterized by widespread inflammation in the lungs that results in flooding of the lungs' microscopic air sacs, which are responsible for the exchange of gases such as oxygen and carbon dioxide with capillaries in the lungs. Additional common findings in ARDS include partial collapse of the lungs (atelectasis) and low levels of oxygen in the blood (hypoxemia).
- the clinical syndrome is associated with pathological findings including pneumonia and diffuse alveolar damage, the latter of which is characterized by diffuse inflammation of lung tissue.
- the triggering insult to the tissue usually results in an initial release of chemical signals and other inflammatory mediators secreted by local epithelial and endothelial cells.
- ARDS impairs the lungs' ability to exchange oxygen and carbon dioxide.
- the underlying mechanism of ARDS involves diffuse injury to cells that form the barrier of the microscopic air sacs of the lungs, surfactant dysfunction, activation of the immune system, and dysfunction of the body's regulation of blood clotting. Diagnosis of ARDS is based on the 2012 Berlin definition:
- ARDS The severity of ARDS is defined by the Berlin definition as:
- ARDS caused by a respiratory viral infection can lead to damage to many organs, including the lungs, heart, and kidneys.
- SARS-CoV-2 infects endothelial cells and also leads to systemic inflammation, causing vasculopathy that affects widespread parts of the body.
- the vasculopathy or other phenomena can cause damage to the heart (myocarditis or arrhythmia), kidneys (acute kidney injury, chronic kidney disease, or renal failure), liver (liver dysfunction), blood vessels (bleeding and blood clots), skin (Kawasaki-like syndrome, rash, hair loss, and urticarial, vesicular, purpuric, and papulosquamous lesions), digestive system (anorexia, nausea, vomiting, diarrhea, and abdominal pain), brain, and nerves (cerebrovascular disease, ataxia, seizure, vision impairment, and nerve pain). Other symptoms include lymphopenia, hypoxia, blood hypercoagulability, multi-organ failure, sepsis, and septic shock.
- Anti-viral compositions include protease inhibitors, nucleoside analogs, antibodies (including monoclonal antibodies, neutralizing antibodies, and/or convalescent plasma), and other types of drugs.
- Administration and Dosing include protease inhibitors, nucleoside analogs, antibodies (including monoclonal antibodies, neutralizing antibodies, and/or convalescent plasma), and other types of drugs.
- compositions provided by the present disclosure can be used for suppressing inflammation in the lungs, thus making them therapeutics for ARDS, ARDS triggered by Covid-19, and ARDS triggered by any viral respiratory disease.
- DA-DKP-containing compositions provided by the present disclosure can be administered to a subject by any suitable route of administration to the lungs, including nasal, intratracheal, bronchial, direct instillation into the lung, inhaled and oral.
- the DA- DKP-containing compositions of the present invention can also be administered enterally (e.g ., oral, sublingual, buccal, rectal), parenterally (e.g., intravenous, intramuscular, subcutaneous), intranasally, by inhalation, vaginally, or by other routes of administration.
- forms for administration of the DA-DKP-containing compositions of the present disclosure include nebulized form, aerosolized form, sprays, drops, and powders.
- the active ingredient may be mixed under sterile conditions with a pharmaceutically-acceptable carrier and with any buffers, excipients or propellants as may be needed to suit a particular composition and/or delivery method.
- the form for administration is a sterile liquid that is administered as a nebulized liquid form or intravenously.
- Aerosol (inhalation) delivery can be performed using methods standard in the art. Carriers suitable for aerosol delivery are described herein. Devices for delivery of aerosolized formulations include, but are not limited to, pressurized metered dose inhalers (MDI), dry powder inhalers (DPI), and metered solution devices (MSI), and include devices that are nebulizers and inhalers.
- MDI pressurized metered dose inhalers
- DPI dry powder inhalers
- MSI metered solution devices
- the amount of a disclosed therapeutic composition that will be effective in the treatment of a particular disorder or condition disclosed herein will depend on the nature of the disorder or condition, and can be determined by clinical techniques. For example, in vitro or in vivo assays may be employed to help identify optimal dose ranges. Effective dosage amounts may vary with the severity of the disease or condition, the route(s) of administration, the duration of the treatment, the identity of any other drugs being administered to the subject, the age, size and species of the subject, the discretion of the prescribing health care provider, and like factors.
- compositions provided by the present disclosure comprise DA-DKP having a concentration range with a lower endpoint of about 10 mM, about 20 pM, about 30 pM, about 40 mM, about 50 mM, about 60 mM, about 70 mM, about 80 mM, about 90 mM, about 100 mM, about 110 mM, about 120 mM, about 130 mM, about 140 mM, about 150 mM, about 160 mM, about 170 mM, about 180 mM, about 190 mM, about 200 mM, about 210 mM, about 220 mM, about 230 mM, about 240 mM, about 240, about 250 mM, about 260 mM, about 270 mM, about 280 mM, about 290 mM, about 300 mM, about 310, about 320 mM, about 330 mM, about 340 mM, about 350 mM, about 360
- an effective amount of DA-DKP in a composition provided by the present disclosure can vary.
- the effective amount comprises a range of amounts having a lower endpoint of about 10 pg, about 15 pg, about 20 pg, about 25 pg, about 30 pg, about 35 pg, about 40 pg, about 45 pg, about 50 pg, about 55 pg, about 60 pg, about 65 pg, about 70 pg, about 75 pg, about 80 pg, about 85 pg, about 90 pg, about 95 pg, about 100 pg, about 110 pg, about 120 pg, about 130 pg, about 140 pg, about 150 pg, about 160 pg, about 170 pg, about 180 pg, about 190 pg, about 200 pg, about 210 pg, about 220 pg, about 230 pg, about 240 pg, about 250 pg, about
- the effective amount comprises a range of amounts having an upper endpoint of about 500 pg, about 490 pg, about 480 pg, about 470 pg, about 460 pg, about 450 pg, about 440 pg, about 430 pg, about 420 pg, about 410 pg, about 400 pg, about 390 pg, about 380 pg, about 370 pg, about 360 pg, about 350 pg, about 340 pg, about 330 pg, about 320 pg, about 310 pg, about 300 pg, about 290 pg, about 280 pg, about 270 pg, about 260 pg, about 250 pg, about 240 pg, about 230 pg, about 220 pg, about 210 pg, about 200 pg, about 190 pg, about 180 pg, about 170 pg, about 160 pg
- compositions can be used with different routes of administration.
- Administration to the lung can involve doses of about 1, about 2, about 3, about 4, about 5, about 6, about 7, about 8, about 9, about 10, about 11, about 12, about 13, about 14, about 15, about 16, about 17, about 18, about 19, or about 20 milliliters.
- Intravenous administration can involve doses of about 50, about 55, about 60, about 65, about 70, about 75, about 80, about 85, about 90, about 95, about 100, about 105, about 110, about 115, about 120, about 125, about 130, about 135, about 140, about 145, about 150, about 155, about 160, about 165, about 170, about 175, about 180, about 185, about 190, about 195, about 200, about 205, about 210, about 215, about 220, about 225, about 230, about 235, about 240, about 245, about 250, about 260, about 270, about 275, about 280, about 290, about 300, about 310, about 320, about 330, about 340, about 350, about 360, about 370, about 375, about 380, about 390, about 400, about 425, about 450, about 475, about 500, about 525, about 550, about 575, about 600, about 650, about 700, about 750
- compositions can be administered 1, 2, 3, 4, 5, 6, 7, 8, or more times per day for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, or 14 days.
- Disclosed compositions can also be administered for 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 weeks, and/or for 4, 5, 6, 7, 8, 9, 10, 11, or 12 months or more.
- the disclosed compositions can be administered once every three months.
- the concentration of DA-DKP in a composition is selected from about 50 mM to about 350 mM and about 110 mM to about 200 mM; the composition also comprises N-acetyl-tryptophan (NAT), caprylic acid, caprylate or combinations thereof each independently in an amount selected from about 1 mM to about 20 mM and about 1 mM to about 4 mM; and the composition is administered as a multi-dose regimen of between 2 and 6 doses administered every 52 weeks and the amount of each dose is between about 2 mL and about 6 mL, in some embodiments each dose is 4 mL.
- NAT N-acetyl-tryptophan
- compositions of the present disclosure comprise any combination of one or more compounds of the present disclosure as an active ingredient in admixture with one or more pharmaceutically-acceptable carriers and, optionally, with one or more other compounds, drugs or other materials.
- the compound is DA-DKP.
- Each carrier is advantageously "acceptable" in the sense of being compatible with the other ingredients of the formulation and not injurious to the subject.
- the compounds of the present disclosure are formulated into pharmaceutically-acceptable dosage forms by conventional methods (see, e.g., Remington’s Pharmaceutical Sciences (Easton, Pa: Mack Pub. Co, 1965. Print; 23 rd Ed. (2020) ISBN: 9780128200070)).
- aqueous and nonaqueous carriers examples include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate.
- polyols such as glycerol, propylene glycol, polyethylene glycol, and the like
- vegetable oils such as olive oil
- injectable organic esters such as ethyl oleate.
- Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.
- compositions may also contain adjuvants such as wetting agents, emulsifying agents and dispersing agents. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like in the compositions. In addition, prolonged absorption of an injectable pharmaceutical form may be brought about by the inclusion of agents which delay absorption such as aluminum monostearate and gelatin.
- the formulations may be presented in unit-dose or multi-dose sealed containers, for example, ampoules and vials, and may be stored in a lyophilized condition requiring only the addition of the sterile liquid carrier, for example water for nebulization, immediately prior to use.
- the sterile liquid carrier for example water for nebulization
- kits comprising the pharmaceutical products of the present disclosure are also provided.
- the kits can comprise a DA-DKP composition formulated for administration to the lung including a nebulized form and/or an aerosolized form.
- the DA-DKP can be prepared as described herein, such as by removing albumin from a solution of a human albumin composition.
- the kits may contain unit-dose or multi-dose sealed containers, for example, ampoules and vials, and may be stored in a lyophilized condition requiring only the addition of the sterile liquid carrier, for example water, immediately prior to use.
- the kits may also be stored in a condition, wherein the contents are ready for direct use or injection.
- compositions of the present disclosure may further comprise N-acetyl- tryptophan (NAT), caprylic acid, caprylate or combinations thereof.
- the compositions comprise NAT.
- the concentration range of each of NAT, caprylic acid, and/or caprylate have a lower endpoint of about 1 mM, about 2 mM, about 3 mM, about 4 mM, about 5 mM, about 6 mM, about 7 mM, about 8 mM, about 9 mM, about 10 mM, about 11 mM, about 12 mM, about 13 mM, about 14 mM, about 15 mM, about 16 mM, about 17 mM, about 18 mM, about 19 mM, or about 20 mM.
- compositions of the present disclosure having NAT, caprylic acid, caprylate or combinations thereof have a concentration range with an upper endpoint of about 40 mM, about 39 mM, about 38 mM, about 37 mM, about 36 mM, about 35 mM, about 34 mM, about 33 mM, about 32 mM, about 31 mM, about 30 mM, about 29 mM, about 28 mM, about 27 mM, about 26 mM, about 25 mM, about 24 mM, about 23 mM, about 22, or about 21 mM.
- the concentration range is about 4 mM to about 20 mM.
- the DA-DKP-containing composition of the present disclosure can also comprise a second drug such as an analgesic (such as lidocaine or paracetamol), an anti inflammatory (such as corticosteroids, such as dexamethasone and betamethasone, non steroid anti-inflammatory drugs (NSAIDs), ibuprofen, naproxen), and/or other suitable drugs.
- analgesic such as lidocaine or paracetamol
- an anti inflammatory such as corticosteroids, such as dexamethasone and betamethasone, non steroid anti-inflammatory drugs (NSAIDs), ibuprofen, naproxen
- NSAIDs non steroid anti-inflammatory drugs
- diketopiperazines such as DA-DKP
- these methods may be employed to synthesize the diketopiperazines of the present disclosure. See, e.g., U.S. Patents Nos. 4,694,081, 5,817,751, 5,990,112, 5,932,579 and 6,555,543, US Patent Application Publication Number 2004/0024180, PCT applications WO 96/00391 and WO 97/48685, and Smith et al., Bioorg. Med. Chem. Letters, 8, 2369- 2374 (1998).
- diketopiperazines such as DA-DKP
- DA-DKP can be prepared by first synthesizing dipeptides.
- the dipeptides can be synthesized by methods well known in the art using L-amino acids, D-amino acids or a combination of D- and L-amino acids.
- solid-phase peptide synthetic methods are employed.
- Dipeptides are also available commercially from numerous sources, including DMI Synthesis Ltd., Edinburgh, UK (custom synthesis), Sigma-Aldrich, St. Louis, MO (primarily custom synthesis), Phoenix Pharmaceuticals, Inc., Belmont, CA (custom synthesis), Fisher Scientific (custom synthesis) and Advanced ChemTech, Louisville, KY.
- the dipeptide is synthesized or purchased, it is cyclized to form a diketopiperazine. This can be accomplished by a variety of techniques including, for example, the method provided in U.S. Patent Application Publication Number 2004/0024180, which describes a method of cyclizing dipeptides. Briefly, the dipeptide is heated in an organic solvent while removing water by distillation.
- the organic solvent is a low-boiling azeotrope with water, such as acetonitrile, allyl alcohol, benzene, benzyl alcohol, n-butanol, 2-butanol, t-butanol, acetic acid butylester, carbon tetrachloride, chlorobenzene chloroform, cyclohexane, 1,2-dichlorethane, diethylacetal, dimethylacetal, acetic acid ethylester, heptane, methylisobutylketone, 3-pentanol, toluene and xylene.
- water such as acetonitrile, allyl alcohol, benzene, benzyl alcohol, n-butanol, 2-butanol, t-butanol, acetic acid butylester, carbon tetrachloride, chlorobenzene chloroform, cyclohexane, 1,2-
- the temperature depends on the reaction speed at which the cyclization takes place and on the type of azeotroping agent used. In some embodiments, the reaction is carried out at 50-200°C, in some embodiments at 80-150°C.
- the pH range in which cyclization takes place can be 2-9, in some embodiments 3-7.
- the dipeptide When one or both of the amino acids of the dipeptide has, or is derivatized to have, a carboxyl group on its side chain (e.g ., aspartic acid or glutamic acid), the dipeptide is cyclized, for example as described in U.S. Patent No. 6,555,543. Briefly, the dipeptide, with the side-chain carboxyl still protected, is heated under neutral conditions. Typically, the dipeptide will be heated at from about 80°C to about 180°C, in some embodiments at about 120°C.
- the solvent will be a neutral solvent.
- the solvent may be an alcohol (such as butanol, methanol, ethanol, and higher alcohols, but not phenol) and an azeotropic co-solvent (such as toluene, benzene, or xylene).
- the alcohol is butan-2-ol
- the azeotropic co-solvent is toluene.
- the heating is continued until the reaction is complete, and such times can be determined empirically.
- the dipeptide will be cyclized by refluxing it for about 8-24 hours, in some embodiments about 18 hours.
- the protecting group is removed from the diketopiperazine, preferably without the use of strong acids (mineral acids, such as sulfuric or hydrochloric acids), strong bases (alkaline bases, such as potassium hydroxide or sodium hydroxide), and/or strong reducing agents (e.g ., lithium aluminum hydride), in order to maintain the chirality of the final compound.
- strong acids mineral acids, such as sulfuric or hydrochloric acids
- strong bases alkaline bases, such as potassium hydroxide or sodium hydroxide
- strong reducing agents e.g ., lithium aluminum hydride
- the compounds described herein encompass all possible enantiomers and stereoisomers including the stereoisomerically pure form (e.g., geometrically pure, enantiomerically pure or diastereomerically pure) and enantiomeric and stereoisomeric mixtures.
- Enantiomeric and stereoisomeric mixtures can be resolved into their component enantiomers or stereoisomers using separation techniques or chiral synthesis techniques.
- Dipeptides made on solid phase resins can be cyclized and released from the resin in one step (see, e.g, U.S. Patent No. 5,817,751).
- resin having an N-alkylated dipeptide attached can be suspended in toluene or toluene/ethanol in the presence of acetic acid (e.g., 1%) or triethylamine (e.g., 4%).
- acetic acid e.g., 1
- triethylamine e.g., 4%
- basic cyclization conditions are utilized for their faster cyclization times.
- diketopiperazines can be made as described below from proteins and peptides.
- diketopiperazines for use in the practice of the present disclosure can be obtained commercially, for example from DMI Synthesis Ltd., Cambridge, UK (custom synthesis).
- the DA-DKP compositions and/or products of the present disclosure can be prepared from solutions containing DA-DKP, including from commercially-available pharmaceutical compositions comprising albumin, such as human serum albumin, using methods such as ultrafiltration, chromatography, size-exclusion chromatography (e.g, Centricon filtration), affinity chromatography (e.g, using a column of beads having attached thereto an antibody or antibodies directed to the desired diketopiperazine(s) or an antibody or antibodies directed to the truncated protein or peptide), anion exchange or cation exchange, sucrose gradient centrifugation, salt precipitation, sonication, or other techniques that will remove some or all of the albumin in the solution.
- albumin such as human serum albumin
- compositions and/or products can be used and incorporated into pharmaceutical compositions as described above.
- the disclosed compositions and/or products can be prepared using an ultrafiltration separation method, whereby a human serum albumin composition can be passed over an ultrafiltration membrane having a molecular weight cut-off that retains the albumin while the DA-DKP passes into the resulting filtrate or fraction.
- This filtrate comprises components having molecular weights selected from less than about 50 kDa, less than about 40 kDa, less than 30 kDa, less than about 20 kDa, less than about 10 kDa, less than about 5 kDa, and less than about 3 kDa.
- the filtrate is a LMWF comprising components having molecular weights less than about 5 kDa.
- This LMWF fraction or filtrate contains DA-DKP which is formed after the dipeptide aspartate-alanine is cleaved from albumin and subsequently cyclized into the diketopiperazine.
- AMPION ® (Ampio Pharmaceuticals, Inc., Englewood, CO USA) is a LMWF ( ⁇ 5 kDa fraction) of human serum albumin (HSA).
- HSA human serum albumin
- AMPION ® can be produced as described herein, for example by filtering commercially available HSA.
- Commercially available HSA is produced by fractionation of blood, for example by the Cohn process or its variations.
- AMPION ® can be produced by filtering commercially available HSA, for example a 5% HSA solution, to remove components above 5 kDa.
- Physiologically-acceptable salts of the DA-DKP of the present disclosure may also be used in the practice of the present disclosure.
- Physiologically-acceptable salts include conventional non-toxic salts, such as salts derived from inorganic acids (such as hydrochloric, hydrobromic, sulfuric, phosphoric, nitric, and the like), organic acids (such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, glutamic, aspartic, benzoic, salicylic, oxalic, ascorbic acid, and the like) or bases (such as the hydroxide, carbonate or bicarbonate of a pharmaceutically-acceptable metal cation or organic cations derived from N,N-dibenzylethylenediamine, D-glucosamine, or ethylenediamine).
- the salts are prepared in a conventional manner, for example by neutralizing the free base form of the compound with an acid.
- methods of treating a viral respiratory disease comprising administration of the disclosed compositions in combination with one or more additional therapeutics for the treatment of the viral respiratory disease.
- the methods relate to the treatment of a viral respiratory disease by administration of two or more therapeutics, each independently formulated to treat the viral respiratory disease.
- the two therapeutics can be administered concurrently or at different time points, via the same route or via different routes.
- the two therapeutics are selected to administer complimentary, combination therapy to a subject suffering from a viral respiratory disease.
- Combination therapy is a treatment modality that combines two or more therapeutic agents.
- the amalgamation of therapeutic agents to treat viral respiratory diseases may enhance efficacy compared to the mono-therapy approach, as the therapeutic agents can be selected to target key pathways in a synergistic and/or additive manner.
- This approach has many benefits, including potentially reducing drug resistance, while simultaneously providing therapeutic benefits.
- the methods comprise administration of a first drug effective to treat a viral respiratory disease, for example COVID-19, and also administering a pharmaceutical composition provided by the present disclosure, comprising DA-DKP, to treat the same viral respiratory disease.
- the methods comprise administration of a first drug before a hyperinflammatory stage of a viral respiratory disease and administering a pharmaceutical composition provided by the present disclosure, comprising DA-DKP, to treat a hyperinflammatory stage of the viral respiratory disease.
- the pharmaceutical composition comprising DA-DKP can be administered before, at, or after onset of the hyperinflammatory stage of the viral respiratory disease.
- the first drug can be one or more antiviral, immune-modifying, anti-depressant, and/or corticosteroid drugs, including combinations thereof.
- the first drug is selected from: Protease inhibitors: Paxlovid (PF-07321332) - a protease inhibitor antiviral therapy, nirmatrelvir (an orally active 3C-like protease inhibitor), ritonavir (protease and cytochrome P450-3A4 (CYP3A4) inhibitor); Nucleoside analogs (or prodrugs thereof): Molnupiravir - a prodrug of the synthetic nucleoside derivative N 4 - hydroxycytidine, Remdesivir - a prodrug of nucleoside analog GS-441524, Favipiravir - a prodrug of purine nucleic acid analog favipiravir-ribofuranosyl-5'-triphosphate; Immune- modifying antibodies: Tocilizumab -
- AMPION ® reduces CL075- and CL307 -induced CXCL10 release from PMA-differentiated THP-1 cells.
- CL075 and CL307 agonists were used to activate TLR7/8 or TLR7, respectively, in PMA-differentiated THP-1 cells. Increases in CXCL10 release indicate progression to hyperinflammation, and decreases indicate inhibition of progression to hyperinflammation.
- AMPION ® was manufactured by Ampio Pharmaceuticals, Inc. (Englewood, CO).
- General cell culture reagents were purchased from ThermoFisher Scientific (Waltham, MA) while X-Vivo 15 serum-free medium was obtained from Lonza (Basel, Switzerland). 0.9% (w/v) sodium chloride was obtained from KD Medical (Columbia, MD).
- GW9662 and CH223191 antagonists were purchased from MilliporeSigma (St. Louis, MO) with 100 mM stock solutions prepared in DMSO and stored at -80°C prior to use.
- CL075 and CL307 were purchased from Invivogen (San Diego, CA) and stock solutions prepared using supplied sterile, pyrogen-free water.
- ELISAs for IPIO/CXCLIO catalog # KAC2361
- All other reagents were obtained from MilliporeSigma (St. Louis, MO) unless otherwise stated.
- Human THP-1 monocytes (ATCC, Manassas, VA) were passaged in RPMI 1640 media supplemented with 10% fetal bovine serum (FBS) and 1% penicillin/ streptomycin (Pen/ Strep) then differentiated using a final concentration of 50 ng/ml phorbol 12-myristate (PMA) in 96-well flat bottom plates seeded at 1 c 10 5 cells per well for 72 hours. Differentiation mediums were then aspirated and replenished as described below.
- FBS fetal bovine serum
- Pen/ Strep penicillin/ streptomycin
- AMPION ® reduces CL075- and CL307 -induced CXCL10 release from Peripheral Blood Mononuclear Cells (PBMC).
- PBMC Peripheral Blood Mononuclear Cells
- AMPION ® was also found to inhibit CXCL10 release from TLR4-activated PBMC. To establish if AMPION ® can influence ex vivo CL075- and CL307-induced CXCL10, these agonists were used to activate TLR7/8 in PBMC cocultures.
- cryopreserved vials obtained from Zen-Bio (Research Triangle Park, NC) were thawed using a Thawstar Automated Cell Thawing System (BioLife Solutions, Bothell, WA) and then transferred dropwise to RPMI 1640 medium containing 10% human AB serum, 1% penicillin-streptomycin (Pen/Strep), and 2 U/mL RNase-free DNase (ThermoScientific).
- Freshly isolated cells were isolated from sodium heparinized whole blood using Polymorphprep (Alere Technologies, Oslo, Norway) and washed with Dulbecco’s phosphate buffer saline.
- the resulting cell suspensions were then centrifuged at 1000 rpm for 10 minutes and working suspensions prepared at 2 x 10 6 cells/mL in X-Vivo 15 or RPMI 1640 supplemented with 20% FBS, 2% Pen/Strep, 1% sodium bicarbonate, 7.5% solution, 1% 100 mM sodium pyruvate, 1% 100X MEM non- essential amino acid solution, and 1% 200 mM L-glutamine.
- PBMC suspensions were added to 96-well tissue culture plates or 100 m ⁇ of the 20% FBS RPMI medium described above was added to THP-1 plate wells.
- the solutions were then mixed with an equal volume of sterile 0.9% sodium chloride or AMPION ® drug solutions and incubated at 37 °C and 5% CO2 for one hour. Stimulation was then achieved by adding CL075 or CL307 to the final concentrations indicated for an additional 24 to 72 hours before subsequent analysis.
- 20 mM (10 mM final) concentration of antagonists were added to mediums prior to addition of saline or drug solutions serially diluted in saline. After the indicated stimulation periods described in these Examples, supernatants were collected for in-house CXCL10 ELISA measurements.
- CXCL10 release induced by CL075 from PBMC is observed 24 hours following exposure (Fig. 2A).
- Fig. 2A an inverse relationship of CXCL10 release to CL075 concentration was observed using the donor cells represented in Fig. 2A, suggesting a biphasic response.
- AMPION ® treatment resulted in significant inhibitions of 93 ⁇ 10%, 55 ⁇ 4%, and 65 ⁇ 2% in CXCL10 release from 5 pg/ml, 2 pg/ml, and 0.5 pg/ml concentrations of agonist, respectively, when corrected for the background chemokine release observed for this donor.
- Percent inhibition in CXCL10 release normalizes 24-hour CL075-induced PMA- differentiated THP-1 and PBMC data
- Fig. 3C demonstrates how normalization within each experiment to percent inhibition in CXCL10 release versus control corrected for observed background chemokine release and transformed the data to a more symmetrical distribution.
- PPARy and AhR antagonists reduce AMPION ® drug potency as measured by CL075- induced CXCL10 release.
- ELISA array analysis demonstrates that AMPION * inhibits the release of a diverse set of pro-inflammatory cytokines and chemokines from CL075-activated, PMA-differentiated 0 THP-1 cells.
- a multi-plex ELISA array was performed to assess the release of 48 cytokines, chemokines, and growth factors, to determine the overall immune response in PMA- differentiated THP-1 cells following CL075 activation and to identify additional drugs effects.
- the ELISA was Eve Technologies (Calgary, Alberta, Canada) human 5 cytokine/chemokine 48-plex Discovery Assay Array (catalog # HD48).
- the top term returned from the Wikipathways library of Enrichr was the ‘COVID-19 adverse outcome pathway’ (WP4891) followed by others such as ‘cytokine responses and inflammatory response’ (WP530) or ‘toll-like receptor signaling’ (WP75) as well as Tung fibrosis’ (WP3624) and ‘lymphoid development/polarization’ (WP3893 and WP4494).
- WP4891 ‘COVID-19 adverse outcome pathway’
- WP530 cytokine responses and inflammatory response
- WP75 toll-like receptor signaling
- WP3624 Tung fibrosis
- WP3893 and WP4494 ‘lymphoid development/polarization’
- IP A Ingenuity Pathway Analysis
- IP A calculated differentially abundant cytokines/chemokines were uploaded, and a ‘Core’ expression analysis based on log ratios for analytes with a p-value ⁇ 0.05 was run using our ‘user dataset’ as reference.
- IPA calculated overlap p-values and z-scores as confidence metrics when predicting canonical pathway associations. For overlap p-values, we considered the - log(pvalue)>1.3 as significant.
- a patient hospitalized due to COVID-19 receives standard of care therapy with the addition of AMPION ® to prevent, reduce the severity or duration of, or treat a hyperinflammatory stage of COVID-19.
- the patient is administered remdesivir, remdesivir plus dexamethasone, or dexamethasone.
- the patient may also be administered one or more other antiviral drugs.
- the patient is administered AMPION ® before, at, or after onset of the hyperinflammatory stage. If the patient experiences rapidly increasing oxygen needs and systemic inflammation, baricitinab, tocilizumab, sarilumab, and/or another immunomodulatory drug may be administered.
- the antiviral drug or drugs inhibit the viral infection, and AMPION ® treats the hyperinflammatory stage of COVID-19.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Veterinary Medicine (AREA)
- Public Health (AREA)
- General Health & Medical Sciences (AREA)
- Medicinal Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Animal Behavior & Ethology (AREA)
- Epidemiology (AREA)
- Virology (AREA)
- Communicable Diseases (AREA)
- Oncology (AREA)
- Organic Chemistry (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- General Chemical & Material Sciences (AREA)
- Molecular Biology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Engineering & Computer Science (AREA)
- Zoology (AREA)
- Gastroenterology & Hepatology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Immunology (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
Abstract
The present disclosure provides methods of treating a TLR7- and/or TLR-mediated disease and also methods of treating disease in a multi-modal treatment regimen. The methods comprise administering an effective amount of a pharmaceutical composition prepared by removing albumin from a solution of a human serum albumin composition and/or comprising a diketopiperazine with amino acid side chains of aspartic acid and alanine (DA-DKP), such as a low molecular weight fraction of human serum albumin.
Description
METHODS OF TREATING VIRAL DISEASES
FIELD
The present disclosure relates to methods of treating TLR7/8-mediated disease, including viral respiratory disease. The methods comprise administering an effective amount of a pharmaceutical composition prepared by removing albumin from a solution of a human serum albumin composition and/or comprising a diketopiperazine with amino acid side chains of aspartic acid and alanine (DA-DKP), such as a low molecular weight fraction of human serum albumin.
BACKGROUND
Toll-like receptors (TLRs) are key pattern recognition complexes that help the immune system respond to pathogens based on unique microbial molecular structures. However, tissue damage can result in the release of cellular components that mimic these molecular motifs and has been shown to play a role in the pathophysiology of diseases through the activation of TLRs.
Treatments that modulate and/or interrupt TLR signaling may prove beneficial for the treatment of diseases, for example when immune cells are activated by antiviral mechanisms that may help drive hyperinflammation in patients with viral infections, especially respiratory viruses. For example, mounting evidence suggests that clinical outcomes vary greatly for coronavirus disease of 2019 (COVID-19) depending on how the immune system responds to severe acute respiratory syndrome coronavirus 2 (SARS-CoV- 2) infection. In its mildest forms, SARS-CoV-2 replication is primarily localized to the upper respiratory tract, with limited innate immune response and low viral burden. However, underlying factors such as genetic polymorphisms, autoantibody development, and intrinsic viral mechanisms have been identified that serve to suppress or delay type I interferon production or activity during the course of infection. The resulting maladaptation in antiviral immunity helps drive progression into more severe stages marked by migration of the virus into the lower respiratory tract, elevated viral loads, and dramatic loss of type II pneumocytes.
As the disease enters these phases, ongoing activation of TLRs leads to concomitant activation of innate immune effector mechanisms, including the production of pro- inflammatory cytokines, and up-regulation of MHC molecules and co-stimulatory signals in
antigen-presenting cells as well as activating natural killer (NK) cells. Upregulation of pro- inflammatory cytokines like CXCL10 become predictive of the clinical course, and approximately 20% of these patients go on to develop acute respiratory distress syndrome (ARDS). Moreover, these critical stages are characterized by increased plasma levels of pro- inflammatory cytokines and chemokines, such as IFNy, CXCL10, IL-Ib, and TNFa, indicative of “cytokine storm” development that can eventually lead to multiorgan failure and death. Thus, it appears that failure of the immune system to mount an appropriate antiviral response early in infection drives a pathology involving excessive tissue damage, CD4+ T-cell helper type 1 (Thl) inflammation, and fibrosis.
Diketopiperazines have been reported to exhibit a variety of biological activities. See, e.g, U.S. Patent Nos. 4,289,759 (immunoregulatory agents), 4,331,595 (immunoregulatory agents), 4,940,709 (PAF antagonists), 5,700,804 (inhibitors of plasminogen activator inhibitor), 5,750,530 (inhibitors of plasminogen activator inhibitor) and 5,990,112 (inhibitors of metalloproteases); PCT publication nos. WO 97/36888 (inhibitors of farnesyl-protein transferase) and WO 99/40931 (treatment of central nervous system injury); EP Patent No. 0043219 (immunoregulatory agents); Japanese patent application nos. 63 290868 (PAF antagonists) and 31 76478 (immunosuppressive agents); and Shimazaki et ak, Chem. Pharm. Bull., 35(8), 3527-3530 (1987) (PAF antagonists), Shimazaki et ak, J. Med. Chem., 30, 1709-1711 (1987) (PAF antagonists), Shimazaki et ak, Lipids, 26(12), 1175-1178 (1991) (PAF antagonists), Yoshida et ak, Prog. Biochem. Pharmacol., 22, 68-80 (1988) (PAF antagonists), Alvarez et ak, J. Antibiotics, 47(11), 1195- 1201 (1994) (inhibitors of calpain).
SUMMARY
Disclosed herein are compositions and methods useful to treat diseases mediated by TLR7 and/or TLR8, which recognize single-stranded (ssRNA) RNA viruses. The disclosed compositions and methods are useful to treat symptoms caused by infection with ssRNA viruses and viruses recognized by TLR7 and/or TLR8. For example, the compositions and methods disclosed herein are useful to treat inflammation caused by infection with a virus, for example a respiratory virus and/or a ssRNA respiratory virus. The compositions and methods disclosed herein are also useful to treat other diseases mediated by TLR7 and/or TLR8, such as lupus and/or lupus nephritis.
Also disclosed herein are compositions and methods useful to treat or prevent one or more symptoms of a viral infection using a biphasic or multi-modal treatment method, including treating inflammation during a hyperinflammatory stage of a TLR7- and/or TLR8- mediated disease. According to such methods, treatment of a patient occurs by administration of a first drug, during the acute phase of the disease, followed by administration of a pharmaceutical composition comprising DA-DKP before, at or after the onset of a hyperinflammatory stage of the disease. In various aspects, upon administration of the pharmaceutical composition the first drug and the pharmaceutical composition are co-administered to the patient. In various aspects, the first drug is an antiviral, an immune- modifying drug, an anti-depressant, a corticosteroid, or combinations thereof, among other things.
In a first aspect, a method of treating one or more symptoms of a viral infection in a patient is provided. The method comprises administering a first drug to the patient prior to onset of a hyperinflammatory stage of the infection and administering a pharmaceutical composition comprising DA-DKP to the patient before, at, or after the onset of the hyperinflammatory stage.
In a second aspect, a method of treating inflammation during a hyperinflammatory stage of a TLR7- and/or TLR8-mediated disease in a patient is disclosed. The method comprises administering a first drug to the patient prior to onset of a hyperinflammatory stage of the disease and administering a pharmaceutical composition comprising DA-DKP to the patient before, at, or after the onset of the hyperinflammatory stage.
In a third aspect, a method of treating or preventing one or more symptoms of a TLR7- and/or TLR8-mediated disease in a patient, by administering a pharmaceutical composition comprising DA-DKP, is disclosed.
In a fourth aspect, a method of treating or preventing inflammation associated with a TLR7- and/or TLR8-mediated disease in a patient, by administering a pharmaceutical composition comprising DA-DKP, is disclosed.
BRIEF DESCRIPTION OF THE DRAWINGS
FIGS. 1A and IB show data demonstrating that AMPION® inhibits CXCL10 release from THP-1 cells. Representative CL075- and CL307-induced CXCL10 chemokine release from PMA-differentiated THP-1 cells cultured in the presence of saline diluent control or AMPION®. Data are presented as mean pg/ml CXCL10 ± STD of three technical
replicates 24 hours (FIG. 1 A) or 72 hours (FIG. IB) post stimulation. * = p-value < 0.05 vs activated saline control by student t-test.
FIGS. 2A and 2B show data demonstrating that AMPION® inhibits CXCL10 release from THP-1 cells. Representative CL075- and CL307-induced CXCL10 chemokine release from PMA-differentiated THP-1 cells cultured in the presence of saline diluent control or AMPION®. Data are presented as mean pg/ml CXCL10 ± STD of three technical replicates 24 hours (FIG. 2A) or 72 hours (FIG. 2B) post stimulation. * = p-value < 0.05 vs activated saline control by student t-test.
FIGS. 3A, 3B, and 3C show data demonstrating that AMPION® inhibits CXCL10 release in THP-1 cells and in Peripheral Blood Mononuclear Cells (PBMC). Box plots for 24-hour 5 pg/ml CL075-induced CXCL10 release and AMPION® percent inhibitions for PMA-differentiated THP-1 cells and PBMC are shown. Data are presented as CL075- induced CXCL10 pg/ml release for both saline- and AMPION®-treatment groups from PMA-differentiated THP-1 (FIG. 3 A) or PBMC (FIG. 3B) as well as percent inhibitions in CXCL10 release observed in the AMPION®-treatment groups calculated versus saline- treated controls (FIG. 3C). Boxes = quartile 1 to 3 boundaries, line = Median, + = Mean. For THP-1, n = 8 independent experiments. For PBMC, n = 9 independent experiments using cells derived from a total of 7 different donors.
FIG. 4 shows data from a relative potency assay of AMPION® inhibition of CXCL10. The data show dose-dependent reduction in CL075-induced CXCL10 release from PMA-differentiated THP-1 by AMPION®. Data presented as regression analysis of percent inhibitions ± STD in 24 hour, CL075-induced CXCL10 release versus AMPION® serially diluted using saline diluent with concentrations listed as nominal in relation to full strength drug product n = 9 for nominal 1 or full-strength product and n = 4 for all other doses.
FIG. 5 shows canonical pathway analysis that demonstrates AMPION® directional regulation of cytokines and chemokines are predicted to inhibit pathways associated with increased inflammation. Log2 fold-changes and p-values of cytokines and chemokines from the 48-plex cytokine array comparing saline-treated versus AMPION®-treated PMA- differentiated THP-1 cells activated with 5 pg/ml CL075 for 24h were uploaded into IPA for canonical pathway analysis. Dark grey = pathway predicted to be activated, z-score > 2,
black = pathway predicted to be inhibited, z-score < -2, white = directional regulation unable to be predicted, z = 0.
DETAILED DESCRIPTION
Definitions
The term “LMWF” refers to a Low Molecular Weight Fraction of human serum albumin (HSA) that is a composition prepared by separation of high molecular weight components from HSA. For example, LMWF can be prepared by filtration of commercially available HSA solutions wherein molecular weight components of more than 3 kDa, more than 5 kDa, more than 10 kDa, more than 20 kDa, more than 30 kDa, more than 40 kDa, or more than 50 kDa are separated from the HSA solution. In some embodiments, “LMWF” refers to a composition prepared by separation of high molecular weight components from HSA by other techniques, including but not limited to ultrafiltration, column chromatography including size exclusion chromatography, affinity chromatography, anion exchange, cation exchange, sucrose gradient centrifugation, salt precipitation, and/or sonication. In some embodiments, LMWF also refers to a composition that includes components of HSA having a molecular weight less than 50 kDa, less than 40 kDa, less than 30 kDa, less than 20 kDa, less than 10 kDa, less than 5000 Da, less than 4000 Da, or less than 3000 Da (corresponding to 50,000 g/mol, 40,000 g/ml, 30,000 g/mol, 20,00 g/mol, 10,000 g/mol, 5,000 g/mol, 4,000 g/mol or 3,000 g/mol, respectively).
As used herein, the terms “patient” and “subject” are interchangeable and generally refer to an animal or a human to which a composition disclosed herein is administered or is to be administered.
As used herein, the term “prodrug” refers to derivatives of a pharmacologically active drug molecule which undergo transformation within the body to produce the pharmacologically active drug, also referred to sometimes as the “parent” drug. Prodrugs are frequently, although not necessarily, pharmacologically inactive until converted to the parent drug.
A “therapeutically effective amount,” “effective amount,” or the like means the amount of a compound or composition that, when administered to a patient for treating a disease, is sufficient to effect such treatment for the disease. The “effective amount” will vary depending on the compound, the disease and its severity and the age, weight, etc., of the patient to be treated.
As used herein, the terms “treat,” “treatment,” “treating,” and derivatives thereof mean to reduce (wholly or partially) the symptoms, duration or severity of a disease. In some embodiments, such terms relate to ameliorating a disease or disorder (/. ., arresting or reducing the development of the disease or at least one of the clinical symptoms thereof). In other embodiments, such terms refer to ameliorating at least one physical parameter, which may or may not be discernible by the patient. In yet other embodiments, such terms refer to inhibiting the disease or disorder, either physically, (e.g., stabilization of a discernible symptom), physiologically, (e.g., stabilization of a physical parameter), or both. In still other embodiments, “treating” or “treatment” refers to delaying the onset of the disease or disorder. In accordance with the present state of the art, treat typically does not mean to cure.
Reference will now be made in detail to particular embodiments of compositions and methods. The disclosed embodiments are not intended to be limiting of the claims.
The present disclosure generally relates to methods of treating a viral respiratory disease, for example a TLR7/8-associated disease such as COVID-19, and/or one or more symptoms thereof. In a first aspect, the methods of treatment relate to the administration of the disclosed compositions as mono-therapeutic agents to treat viral respiratory diseases. In a second aspect, the methods of treatment relate to the administration of the disclosed compositions in combination with one or more additional therapeutics for the treatment of a viral respiratory disease.
With respect to the first aspect, the methods comprise administering an effective amount of a pharmaceutical composition comprising aspartyl-alanyl diketopiperazine (DA- DKP) to a subject having a need thereof. In various aspects, the pharmaceutical composition can be prepared by removing albumin from a solution of a human serum albumin composition. The pharmaceutical composition can also include N-acetyl-tryptophan (NAT), caprylic acid, caprylate or combinations thereof. In some embodiments, the pharmaceutical composition can comprise AMPION® (defined herein).
With respect to the second aspect, the methods comprise administering an effective amount of a pharmaceutical composition comprising DA-DKP before, during, or after the initiation of a hyperinflammatory stage of viral disease to a subject having a need thereof. The pharmaceutical composition can be prepared as noted above, by removing albumin from a solution of a human serum albumin composition. The pharmaceutical composition
can also include N-acetyl-tryptophan (NAT), caprylic acid, caprylate or combinations thereof. In some embodiments, the pharmaceutical composition can comprise AMPION® (defined herein).
With further respect to the second aspect, in some embodiments the present disclosure generally relates to methods of treating one or more respiratory viral diseases by administering an effective amount of a pharmaceutical composition comprising DA-DKP before a hyperinflammatory stage of respiratory viral disease to a subject having a need thereof. The pharmaceutical composition can be prepared as noted above, by removing albumin from a solution of a human serum albumin composition. The pharmaceutical composition can also include N-acetyl-tryptophan (NAT), caprylic acid, caprylate or combinations thereof. In some embodiments, the pharmaceutical composition can comprise AMPION® (defined herein).
In one embodiment, the present disclosure relates to methods of treating COVID-19 by administering an effective amount of a pharmaceutical composition comprising DA- DKP before a hyperinflammatory stage of COVID-19 to a subject having a need thereof. The pharmaceutical composition can be prepared as noted above, by removing albumin from a solution of a human serum albumin composition. The pharmaceutical composition can also include N-acetyl-tryptophan (NAT), caprylic acid, caprylate or combinations thereof. In some embodiments, the pharmaceutical composition can comprise AMPION® (defined herein).
In other embodiments, the present disclosure relates to methods of treating viral diseases by administering an effective amount of a pharmaceutical composition comprising DA-DKP during a hyperinflammatory stage of viral disease to a subject having a need thereof. The pharmaceutical composition can be prepared as noted above, by removing albumin from a solution of a human serum albumin composition. The pharmaceutical composition can also include N-acetyl-tryptophan (NAT), caprylic acid, caprylate or combinations thereof. In some embodiments, the pharmaceutical composition can comprise AMPION® (defined herein).
In one embodiment, the present disclosure relates to methods of treating COVID-19 by administering an effective amount of a pharmaceutical composition comprising DA- DKP during a hyperinflammatory stage of COVID-19 to a subject having a need thereof. The pharmaceutical composition can be prepared as noted above, by removing albumin
from a solution of a human serum albumin composition. The pharmaceutical composition can also include N-acetyl-tryptophan (NAT), caprylic acid, caprylate or combinations thereof. In some embodiments, the pharmaceutical composition can comprise AMPION® (defined herein).
In other embodiments, the present disclosure relates to methods of treating viral diseases by administering an effective amount of a pharmaceutical composition comprising DA-DKP to a subject after a hyperinflammatory stage of viral disease has begun. The pharmaceutical composition can be prepared as noted above, by removing albumin from a solution of a human serum albumin composition. The pharmaceutical composition can also include N-acetyl-tryptophan (NAT), caprylic acid, caprylate or combinations thereof. In some embodiments, the pharmaceutical composition can comprise AMPION® (defined herein).
In one embodiment, the present disclosure relates to methods of treating COVID-19 by administering an effective amount of a pharmaceutical composition comprising DA- DKP to a subject after a hyperinflammatory stage of COVID-19 has begun. The pharmaceutical composition can be prepared as noted above, by removing albumin from a solution of a human serum albumin composition. The pharmaceutical composition can also include N-acetyl-tryptophan (NAT), caprylic acid, caprylate or combinations thereof. In some embodiments, the pharmaceutical composition can comprise AMPION® (defined herein).
DA-DKP has multiple anti-inflammatory and immune modulating effects including inhibition of multiple pro-inflammatory cytokines, chemokines and signaling molecules at the transcription level, inhibition of the migration and adhesion of T-cells and monocytes, activity at the G-coupled protein receptor level, activity on actin-dependent cytoskeletal events, and reduction in vascular permeability and inhibition of inflammation induced by platelet activating factor, among other effects. DA-DKP can be used to treat lung inflammatory diseases, inflammation related to viral respiratory diseases, and/or to prevent lung inflammation and lung inflammatory diseases related to viral respiratory diseases. DA- DKP can also be used to prevent, reduce in severity and/or duration, and/or otherwise treat cytokine storm or cytokine release syndrome. Cytokine storm or cytokine release syndrome is a systemic inflammatory syndrome characterized by elevated levels of circulating cytokines and immune cell hyperactivation.
Innate immune system and Toll-like Receptors
The innate immune response plays a vital role in the detection of infection and linking acute responses to the adaptive branch. Toll-like receptors (TLR) are one of the primary mechanisms by which innate immune cells detect and acutely respond to microbial infection.
Toll-like receptors (TLRs) recognize specific molecular patterns present in molecules that are broadly shared by pathogens, but are structurally distinct from host molecules. The human genome includes 10 known TLRs. The ligands for these receptors are highly conserved microbial molecules such as lipopolysaccharides (LPS) (recognized by TLR4), lipopeptides (TLR2 in combination with TLR1 or TLR6), flagellin (TLR5), single stranded RNA (TLR7 and TLR8), double stranded RNA (TLR3), CpG motif- containing DNA (recognized by TLR9), and profilin present on uropathogenic bacteria (TLR 11). TLR1, -2, -4, -5, and -6 respond to extracellular stimuli, while TLR3, -7, -8 and -9 respond to intracytoplasmic pathogen-associated molecular patterns (PAMPs, conserved microbial small molecular motifs). The activation of TLRs by their cognate ligands leads to activation of innate immune effector mechanisms, including the production of pro- inflammatory cytokines, and up-regulation of MHC molecules and co-stimulatory signals in antigen-presenting cells as well as activating natural killer (NK) cells. The consequence of activation of the innate immune system mobilizes and amplifies specific adaptive immune responses involving both T- and B-cell effector functions.
Toll-like receptors -7/-8 are innate immune receptors present in the endosomal compartment that are activated by single-stranded RNA (ssRNA) molecules of viral as well as nonviral origin, inducing the production of inflammatory cytokines necessary for the development of adaptive immunity.
TLR7 is an endosomal sensor that can discriminate non-self, single-stranded ribonucleic acid sequences to trigger an anti-viral response. TLR 7 serves to detect non-self, single stranded ribonucleic acid, and trigger anti-viral immunity. However, dysregulation of TLR7 signaling has been linked to the pathology of inflammatory diseases that present with cytokine storms and acute kidney injury such as sepsis and lupus.
TLR7 in Disease
TLR7 is linked to plasma cytokine storm development, acute kidney injury, and mortality. Cytokine storm is a systemic inflammatory syndrome characterized by high levels
of circulating cytokines and hyperactivation of the immune system. This can lead to multiple organ failure and even death. A cytokine storm, or cytokine storm syndrome, can be caused by many infectious and non-infectious etiologies. For example, cytokine storm syndrome can be caused by bacterial and/or viral infection, such as infection by the coronavirus SARS- CoV-2, which causes the disease COVID-19. Cytokine storm symptoms include fever, fatigue, nausea, systemic inflammation, hyperferritinemia, hemodynamic instability, organ failure, and death. Cytokine storm can damage the heart, kidneys, lungs, liver, central nervous system, and other organs, for example by causing cardiac arrest, myocarditis, myocardial injury, cardiomyopathy, arrhythmia, acute kidney injury, proteinuria, and acute respiratory distress syndrome. Furthermore, infection, for example by SARS-CoV-2, can lead to sepsis, which is associated with cytokine storm. Severe COVID-19 patients exhibit cytokine storms associated with acute kidney injury and renal dysfunction. Cytokine storm can be characterized by elevated levels of cytokines including IL-Ib, IL-7, IL-8, IL-9, IL- 10, FGF, G-CSF, GM-CSF, IFN-g, IP-10, MCP-1, MPMA, MIP1-B, PDGF, TNF-a, and VEGF.
TLR7 is also linked to the induction of serum autoantibody products that accelerate glomerulonephritis, exacerbate macrophage activation syndrome, and result in rapid mortality in murine lupus models. Lupus nephritis is linked to TLR7. Lupus nephritis is an autoimmune disease caused in part by the recognition of kidney cells by autoantibodies.
In some embodiments, compositions provided by the present disclosure are useful for the treatment of lupus, including lupus nephritis. Such embodiments are exemplified by methods that can comprise administering an effective amount of a pharmaceutical composition comprising aspartyl-alanyl diketopiperazine (DA-DKP) to a subject having a need thereof. The pharmaceutical composition can be prepared by removing albumin from a solution of a human serum albumin composition. The pharmaceutical composition can also include N-acetyl-tryptophan (NAT), caprylic acid, caprylate or combinations thereof. In some embodiments, the pharmaceutical composition can comprise AMPION® (defined herein).
TLR7/8 recognize single strand RNA (ssRNA) viruses, and inflammation caused by any single strand RNA virus can thus be treated with various aspects of the disclosure. Suitable ssRNA viruses include Coronaviridae ( e.g ., SARS-CoV, MERS, SARS-CoV-2, and toroviruses), Hepeviridae (e.g., Hepatitis E), Caliciviridae (e.g., Norovirus, formerly
Norwalk virus, and Sapporo virus), Togaviridae ( e.g ., Alphavirus viral diseases (e.g., Chikungunya, Eastern equine encephalomyelitis virus, Getah virus, Mayaro virus, Mucambo virus, O’nyong’nyong virus, Ross river virus, Barmah forest virus, Sagiyama virus, Semliki forest virus, Sindbis virus, Tonate virus, Venezuelan equine encephalomyelitis virus, Western equine encephalomyelitis virus) and Rubivirus (e.g., Rubella virus)), Flaviviridae (e.g., Hepacivirus (e.g., Hepatitis C), Flavivirus (e.g., Dengue virus types 1-4, Hepatitis G virus, Japanese B encephalitis virus, Murray Valley encephalitis virus, Rocio virus, Spondweni virus, St Louis encephalitis, Wesselsbron, West Nile virus (West Nile fever), and Yellow fever virus (yellow fever)), viruses in the Tick- borne virus group (e.g., Absettarov, Hanzalova, Hypr, Kumlinge, Kyasanur forest disease, Louping ill (tick-borne encephalitis), Negishi, Omsk, Powassan (tick-borne encephalitis), Langat (tick-borne encephalitis), Russian spring summer encephalitis), and the Hepatitis G virus group (e.g., Hepatitis G virus)), Picornaviridae (e.g., Enterovirus (e.g., Coxsackievirus, Echovirus, Poliovirus, Enterovirus 68-109, and Rhinovirus A and B), Hepatovirus (e.g., Hepatitis A, alternately human enterovirus type 72), Astroviridae (e.g., Astrovirus species), Mononegavirales (e.g., Henipavirus (e.g., Hendra virus and Nipah virus), Rubulavirus (e.g., Mumps virus and Parainfluenza types 2, 4a and 4b), Morbillivirus, (e.g., Measles virus), Avulavirus(e.g·., Newcastle disease virus), Metapneumovirus, Pneumovirus (e.g., Respiratory syncytial virus), and Parainfluenza Types 1 to 4), Rhabdoviridae (e.g., Lyssavirus (e.g., Duvenhage and Rabies virus), Retroviridae (e.g., Human T-cell lymphotropic viruses) and Lentivirus (e.g., Human Immunodeficiency virus)), Arenaviridae (e.g., Arenavirus (e.g., lymphocytic choriomeningitis virus)), Bunyaviridae (e.g., Bunyavirus (e.g., Bunyamwera virus), Hantavirus (e.g., Hantaan virus), Nairovirus (e.g., Nairobi sheep disease virus, and Phlebovirus (e.g., sandfly fever Sicilian virus)), and Orthomyxoviridae (e.g., Influenza viruses (e.g., influenza viruses A,B, C, and D) and Thogoto-like viruses (e.g., Thogoto virus)). Diseases mediated by TLR7 and/or TLR8 are diseases caused by infection with ssRNA viruses recognized by TLR7 and/or TLR8, such as inflammation caused by a ssRNA virus, as well as lupus and lupus nephritis.
Inflammation in viral disease
A complication of disease caused by viral infection is overactive immune response, leading to excessive inflammation. This can lead to development of a hyperinflammatory
stage of infection that is characterized by cytokine storm, acute lung injury (ALI), and/or acute respiratory distress syndrome (ARDS). Many viruses can cause excessive inflammation, cytokine storm, ALI, and/or ARDS, including respiratory viruses, Herpes simplex viruses, and cytomegaloviruses. Certain respiratory viruses are particularly likely to cause excessive inflammation and even ARDS, such as viruses that cause community- acquired pneumonia and/or pandemics. Respiratory viruses that are especially likely to cause ARDS include influenza viruses, coronaviruses, rhinoviruses, respiratory syncytial viruses, parainfluenzaviruses, human metapneumoviruses, and adenoviruses. These include influenza A viruses (including H1N1 and H3N2 strains), influenza B viruses, Severe Acute Respiratory Distress Syndrome coronavirus (SARS-CoV), Middle East Respiratory Syndrome coronavirus (MERS-CoV), Severe Acute Respiratory Distress Syndrome coronavirus 2 (SARS-CoV-2), and human adenovirus B21 infections (HAdV-B21). Other emergent viruses, especially those that cause community-acquired pneumonia and/or pandemics, are likely to also cause ARDS. In various aspects, compositions provided by the present disclosure are useful to treat ARDS caused by any virus.
Viral Respiratory Diseases
A viral respiratory disease is an illness caused by a virus that affects the respiratory tract. Such viral respiratory diseases can include Severe Acute Respiratory Distress Syndrome (SARS), Middle East Respiratory Syndrome (MERS), COVID-19, and viral infections associated with asthma, pneumonia, bronchitis and/or tuberculosis. Viruses that can cause one or more viral respiratory diseases include coronaviruses, influenza viruses, respiratory syncytial virus (RSV), parainfluenza viruses, and respiratory adenoviruses. Coronaviruses include SARS-Coronavirus-2 (SARS-CoV-2), SARS-associated coronavirus (SARS-CoV), and Middle East Respiratory Syndrome Coronavirus (MERS- CoV). Coronavirus infections and other viral infections can cause acute respiratory distress syndrome (ARDS), acute lung injury (ALI), interstitial lung disease, pulmonary fibrosis, pneumonia, and reactive airway disease syndrome. Coronavirus infections and other viral infections can cause inflammation in tissues such as lung, brain, heart, kidney, blood vessel, skin, and nerve. Coronavirus infections and other viral infections can cause symptoms such as fatigue, shortness of breath or difficulty breathing, low exercise tolerance, low blood oxygen saturation, cough, sore throat, stuffy or runny nose, joint pain, chest pain, tightness,
or discomfort, muscle pain, muscle weakness, fever, heart palpitations, difficulty thinking and/or concentrating, and depression.
COVID-19 infection is a respiratory illness caused by the novel coronavirus SARS- COV-2 and has been classified as a pandemic with no known cure to date. COVID-19 is detected and diagnosed with a laboratory test. The primary symptoms of COVID-19 infection include mild symptoms such as fever, cough, chills, muscle pain, headache, gastrointestinal symptoms, and the loss of taste or smell. Once infected, the virus moves down a patient’s respiratory tract, where the lungs may become inflamed, making breathing difficult and sometimes requiring supplemental oxygen in the more severe cases of the disease.
Respiratory symptoms after a COVID-19 infection include shortness of breath, cough, chest discomfort, low exercise tolerance and low oxygen saturation, all of which point to potential inflammation-related complication sequelae. Infiltrating or resident cells in the immune system (e.g., macrophages, peripheral blood mononuclear cells, etc.) may be responsible for the development of these respiratory long-term consequences. Chronic or prolonged inflammation of the lungs may be responsible for a myriad of respiratory signs and symptoms experienced by patients after a COVID-19 infection. Chest x-rays and CT scans reveal disturbing patterns of perhaps extensive fibrosis and potential loss of elasticity and oxygen diffusion capacity.
The SARS-Cov-2 virus transmits through the respiratory system and can cause a severe dysregulation of the immune response and damage in the lungs. The hyperinflammatory state is thought to lead to prolonged clinical complications, and treatment with immunomodulators at this later point in the disease can be more effective than anti-viral treatment.
Inflammation associated with COVID-19 may trigger even more severe complications including pneumonia, acute lung injury (ALI) and/or acute respiratory distress syndrome (ARDS), which is a leading cause of mortality in COVID-19. ARDS is associated with widespread inflammation in the lungs. The underlying mechanism of ARDS involves diffuse injury to cells which form the barrier of the microscopic air sacs (alveoli) of the lung, surfactant dysfunction, and activation of the immune system. The fluid accumulation in the lungs associated with ARDS is partially explained by vascular leakage due to inflammation.
An important aspect of ARDS, triggered by COVID-19, is an initial release of chemical signals and other inflammatory mediators secreted by lung epithelial and endothelial cells. Neutrophils and some T-lymphocytes migrate into the inflamed lung tissue and contribute to the amplification/deterioration of ARDS. A decrease in the production of lipid mediators of inflammation (prostaglandins) may impair the resolution of inflammation associated with ARDS (see Fukunaga, et. al., Cyclooxygenase 2 Plays a Pivotal Role in the Resolution of Acute Lung Injury. Journal of Immunology 2005; 174:5033-5039.; Gao et al J Immunol 2017; 199:2043-2054).
The World Health Organization (WHO)’s Clinical Care for Severe Acute Respiratory Infection: COVID-19 Adaptation recommends early intervention with supplemental oxygen for COVID-19 patients with low blood oxygen saturation (Sp02) beginning with the least invasive modality possible ( e.g . hand-held oxygen source) and moving to more invasive modalities (e.g. bilevel positive airway pressure [BiPAP] and/or non-invasive ventilation (NIV)) as severity increases. Treatment during early intervention for COVID-19 patients with respiratory distress requires monitoring of respiratory function with treatment responsive to disease progression. The CDC recommends following the guidelines for treatment of COVID-19 patients with hypoxia in Surviving Sepsis Campaign: Guidelines on the Management of Critically Ill Adults with COVID-19.
Patients who fail to respond to less-invasive treatment of any respiratory viral infection are at a high risk of developing ARDS, a rapidly progressive disease characterized by widespread inflammation in the lungs that results in flooding of the lungs' microscopic air sacs, which are responsible for the exchange of gases such as oxygen and carbon dioxide with capillaries in the lungs. Additional common findings in ARDS include partial collapse of the lungs (atelectasis) and low levels of oxygen in the blood (hypoxemia). The clinical syndrome is associated with pathological findings including pneumonia and diffuse alveolar damage, the latter of which is characterized by diffuse inflammation of lung tissue. The triggering insult to the tissue usually results in an initial release of chemical signals and other inflammatory mediators secreted by local epithelial and endothelial cells.
ARDS impairs the lungs' ability to exchange oxygen and carbon dioxide. The underlying mechanism of ARDS involves diffuse injury to cells that form the barrier of the microscopic air sacs of the lungs, surfactant dysfunction, activation of the immune system, and dysfunction of the body's regulation of blood clotting.
Diagnosis of ARDS is based on the 2012 Berlin definition:
• lung injury of acute onset, within 1 week of an apparent clinical insult and with progression of respiratory symptoms
• bilateral opacities on chest imaging (chest radiograph or CT) not explained by other lung pathology (e.g. effusion, lobar/lung collapse, or nodules)
• respiratory failure not explained by heart failure or volume overload
• decreased ratio of partial pressure arterial oxygen (PaCk) to fraction of inspired oxygen (FiCk) of less than or equal to 300 mm Hg despite a positive end-expiratory pressure (PEEP) of more than 5 cm EbO.
The severity of ARDS is defined by the Berlin definition as:
• mild ARDS: 201 - 300 mmHg (< 39.9 kPa)
• moderate ARDS: 101 - 200 mmHg (< 26.6 kPa)
• severe ARDS: < 100 mmHg (< 13.3 kPa)
There are no approved treatments for ARDS, and standard of care (SOC) is supportive management.
ARDS caused by a respiratory viral infection, for example a SARS-CoV-2 infection, can lead to damage to many organs, including the lungs, heart, and kidneys. SARS-CoV-2 infects endothelial cells and also leads to systemic inflammation, causing vasculopathy that affects widespread parts of the body. The vasculopathy or other phenomena can cause damage to the heart (myocarditis or arrhythmia), kidneys (acute kidney injury, chronic kidney disease, or renal failure), liver (liver dysfunction), blood vessels (bleeding and blood clots), skin (Kawasaki-like syndrome, rash, hair loss, and urticarial, vesicular, purpuric, and papulosquamous lesions), digestive system (anorexia, nausea, vomiting, diarrhea, and abdominal pain), brain, and nerves (cerebrovascular disease, ataxia, seizure, vision impairment, and nerve pain). Other symptoms include lymphopenia, hypoxia, blood hypercoagulability, multi-organ failure, sepsis, and septic shock.
Conventional pharmaceutical therapies for viral respiratory disease include anti -viral compositions. Anti-viral compositions include protease inhibitors, nucleoside analogs, antibodies (including monoclonal antibodies, neutralizing antibodies, and/or convalescent plasma), and other types of drugs.
Administration and Dosing
Compositions provided by the present disclosure can be used for suppressing inflammation in the lungs, thus making them therapeutics for ARDS, ARDS triggered by Covid-19, and ARDS triggered by any viral respiratory disease.
DA-DKP-containing compositions provided by the present disclosure can be administered to a subject by any suitable route of administration to the lungs, including nasal, intratracheal, bronchial, direct instillation into the lung, inhaled and oral. The DA- DKP-containing compositions of the present invention can also be administered enterally ( e.g ., oral, sublingual, buccal, rectal), parenterally (e.g., intravenous, intramuscular, subcutaneous), intranasally, by inhalation, vaginally, or by other routes of administration.
In any of the methods and compositions disclosed herein, forms for administration of the DA-DKP-containing compositions of the present disclosure include nebulized form, aerosolized form, sprays, drops, and powders. The active ingredient may be mixed under sterile conditions with a pharmaceutically-acceptable carrier and with any buffers, excipients or propellants as may be needed to suit a particular composition and/or delivery method. In some embodiments, the form for administration is a sterile liquid that is administered as a nebulized liquid form or intravenously.
Aerosol (inhalation) delivery can be performed using methods standard in the art. Carriers suitable for aerosol delivery are described herein. Devices for delivery of aerosolized formulations include, but are not limited to, pressurized metered dose inhalers (MDI), dry powder inhalers (DPI), and metered solution devices (MSI), and include devices that are nebulizers and inhalers.
The amount of a disclosed therapeutic composition that will be effective in the treatment of a particular disorder or condition disclosed herein will depend on the nature of the disorder or condition, and can be determined by clinical techniques. For example, in vitro or in vivo assays may be employed to help identify optimal dose ranges. Effective dosage amounts may vary with the severity of the disease or condition, the route(s) of administration, the duration of the treatment, the identity of any other drugs being administered to the subject, the age, size and species of the subject, the discretion of the prescribing health care provider, and like factors.
Compositions provided by the present disclosure comprise DA-DKP having a concentration range with a lower endpoint of about 10 mM, about 20 pM, about 30 pM,
about 40 mM, about 50 mM, about 60 mM, about 70 mM, about 80 mM, about 90 mM, about 100 mM, about 110 mM, about 120 mM, about 130 mM, about 140 mM, about 150 mM, about 160 mM, about 170 mM, about 180 mM, about 190 mM, about 200 mM, about 210 mM, about 220 mM, about 230 mM, about 240 mM, about 240, about 250 mM, about 260 mM, about 270 mM, about 280 mM, about 290 mM, about 300 mM, about 310, about 320 mM, about 330 mM, about 340 mM, about 350 mM, about 360 mM, about 370 mM, about 380 mM, about 390 mM, or about 400 mM; such compositions also comprise DA-DKP having a concentration range with an upper endpoint of about 600 mM, about 580 mM, about 570 mM, about 560 mM, about 550 mM, about 540 mM, about 530 mM, about 520 mM, about 510 mM, about 500 mM, about 490 mM, about 480 mM, about 470 mM, about 460 mM, about 450 mM, about 440 mM, about 430 mM, about 420 mM, about 410 mM, about 400 mM, about 390 mM, about 380 mM, about 370 mM, about 360 mM, about 350, about 340 mM, about 330 mM, about 320 mM, about 310 mM, about 300 mM, about 290 mM, about 280, about 270 mM, about 260 mM, about 250 mM, about 240 mM, about 230 mM, about 220 mM, about 210 mM, or about 200 mM.
An effective amount of DA-DKP in a composition provided by the present disclosure can vary. In some embodiments, the effective amount comprises a range of amounts having a lower endpoint of about 10 pg, about 15 pg, about 20 pg, about 25 pg, about 30 pg, about 35 pg, about 40 pg, about 45 pg, about 50 pg, about 55 pg, about 60 pg, about 65 pg, about 70 pg, about 75 pg, about 80 pg, about 85 pg, about 90 pg, about 95 pg, about 100 pg, about 110 pg, about 120 pg, about 130 pg, about 140 pg, about 150 pg, about 160 pg, about 170 pg, about 180 pg, about 190 pg, about 200 pg, about 210 pg, about 220 pg, about 230 pg, about 240 pg, about 250 pg, about 260 pg, about 270 pg, about 280 pg, about 290 pg, about 300 pg, about 310 pg, about 320 pg, about 330 pg, about 340 pg, about 350 pg, about 360 pg, about 370 pg, about 380 pg, about 390 pg, about 400 pg, about 425 pg, about 450 pg, about 475 pg or about 500 pg. In some embodiments, the effective amount comprises a range of amounts having an upper endpoint of about 500 pg, about 490 pg, about 480 pg, about 470 pg, about 460 pg, about 450 pg, about 440 pg, about 430 pg, about 420 pg, about 410 pg, about 400 pg, about 390 pg, about 380 pg, about 370 pg, about 360 pg, about 350 pg, about 340 pg, about 330 pg, about 320 pg, about 310 pg, about 300 pg, about 290 pg, about 280 pg, about 270 pg, about 260 pg, about 250 pg, about 240 pg, about 230 pg, about 220 pg, about 210 pg, about 200 pg, about 190 pg, about 180 pg, about 170
pg, about 160 pg, about 150 pg, about 140 pg, about 130 pg, about 120 pg, about 110 pg, about 100 pg, about 90 pg, about 80 pg, about 70 pg, about 60 pg, about 50 pg, about 40 pg, about 30 pg, or about 20 pg.
Different doses of the disclosed compositions can be used with different routes of administration. Administration to the lung, for example by nebulizer, can involve doses of about 1, about 2, about 3, about 4, about 5, about 6, about 7, about 8, about 9, about 10, about 11, about 12, about 13, about 14, about 15, about 16, about 17, about 18, about 19, or about 20 milliliters. Intravenous administration can involve doses of about 50, about 55, about 60, about 65, about 70, about 75, about 80, about 85, about 90, about 95, about 100, about 105, about 110, about 115, about 120, about 125, about 130, about 135, about 140, about 145, about 150, about 155, about 160, about 165, about 170, about 175, about 180, about 185, about 190, about 195, about 200, about 205, about 210, about 215, about 220, about 225, about 230, about 235, about 240, about 245, about 250, about 260, about 270, about 275, about 280, about 290, about 300, about 310, about 320, about 330, about 340, about 350, about 360, about 370, about 375, about 380, about 390, about 400, about 425, about 450, about 475, about 500, about 525, about 550, about 575, about 600, about 650, about 700, about 750, about 800, about 850, about 900, about 950, about 1000, about 1100, about 1200, about 1300, about 1400, or about 1500 milliliters (or cubic centimeters). The disclosed compositions can be administered 1, 2, 3, 4, 5, 6, 7, 8, or more times per day for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, or 14 days. Disclosed compositions can also be administered for 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 weeks, and/or for 4, 5, 6, 7, 8, 9, 10, 11, or 12 months or more. In some embodiments, the disclosed compositions can be administered once every three months.
In some embodiments, the concentration of DA-DKP in a composition is selected from about 50 mM to about 350 mM and about 110 mM to about 200 mM; the composition also comprises N-acetyl-tryptophan (NAT), caprylic acid, caprylate or combinations thereof each independently in an amount selected from about 1 mM to about 20 mM and about 1 mM to about 4 mM; and the composition is administered as a multi-dose regimen of between 2 and 6 doses administered every 52 weeks and the amount of each dose is between about 2 mL and about 6 mL, in some embodiments each dose is 4 mL.
While it is possible for a compound of the present disclosure ( e.g ., DA-DKP, N- acetyl -tryptophan (NAT), caprylic acid, caprylate) to be administered alone, in various
aspects the disclosed compounds are administered together as a pharmaceutical composition. The pharmaceutical compositions of the present disclosure comprise any combination of one or more compounds of the present disclosure as an active ingredient in admixture with one or more pharmaceutically-acceptable carriers and, optionally, with one or more other compounds, drugs or other materials. In some embodiments, the compound is DA-DKP. Each carrier is advantageously "acceptable" in the sense of being compatible with the other ingredients of the formulation and not injurious to the subject. Regardless of the route of administration selected, in embodiments the compounds of the present disclosure are formulated into pharmaceutically-acceptable dosage forms by conventional methods (see, e.g., Remington’s Pharmaceutical Sciences (Easton, Pa: Mack Pub. Co, 1965. Print; 23rd Ed. (2020) ISBN: 9780128200070)).
Examples of suitable aqueous and nonaqueous carriers which may be employed in the pharmaceutical compositions of the present disclosure include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate. Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.
The compositions may also contain adjuvants such as wetting agents, emulsifying agents and dispersing agents. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like in the compositions. In addition, prolonged absorption of an injectable pharmaceutical form may be brought about by the inclusion of agents which delay absorption such as aluminum monostearate and gelatin.
The formulations may be presented in unit-dose or multi-dose sealed containers, for example, ampoules and vials, and may be stored in a lyophilized condition requiring only the addition of the sterile liquid carrier, for example water for nebulization, immediately prior to use.
Kits comprising the pharmaceutical products of the present disclosure are also provided. The kits can comprise a DA-DKP composition formulated for administration to the lung including a nebulized form and/or an aerosolized form. The DA-DKP can be prepared as described herein, such as by removing albumin from a solution of a human albumin composition. The kits may contain unit-dose or multi-dose sealed containers, for
example, ampoules and vials, and may be stored in a lyophilized condition requiring only the addition of the sterile liquid carrier, for example water, immediately prior to use. The kits may also be stored in a condition, wherein the contents are ready for direct use or injection.
The compositions of the present disclosure may further comprise N-acetyl- tryptophan (NAT), caprylic acid, caprylate or combinations thereof. In some embodiments, the compositions comprise NAT. In those embodiments where the disclosed compositions have NAT, caprylic acid, caprylate or combinations thereof, the concentration range of each of NAT, caprylic acid, and/or caprylate have a lower endpoint of about 1 mM, about 2 mM, about 3 mM, about 4 mM, about 5 mM, about 6 mM, about 7 mM, about 8 mM, about 9 mM, about 10 mM, about 11 mM, about 12 mM, about 13 mM, about 14 mM, about 15 mM, about 16 mM, about 17 mM, about 18 mM, about 19 mM, or about 20 mM. In addition, compositions of the present disclosure having NAT, caprylic acid, caprylate or combinations thereof have a concentration range with an upper endpoint of about 40 mM, about 39 mM, about 38 mM, about 37 mM, about 36 mM, about 35 mM, about 34 mM, about 33 mM, about 32 mM, about 31 mM, about 30 mM, about 29 mM, about 28 mM, about 27 mM, about 26 mM, about 25 mM, about 24 mM, about 23 mM, about 22, or about 21 mM. In some embodiments, the concentration range is about 4 mM to about 20 mM.
In addition, the DA-DKP-containing composition of the present disclosure can also comprise a second drug such as an analgesic (such as lidocaine or paracetamol), an anti inflammatory (such as corticosteroids, such as dexamethasone and betamethasone, non steroid anti-inflammatory drugs (NSAIDs), ibuprofen, naproxen), and/or other suitable drugs.
Synthesis
Methods of making diketopiperazines, such as DA-DKP, are well known in the art, and these methods may be employed to synthesize the diketopiperazines of the present disclosure. See, e.g., U.S. Patents Nos. 4,694,081, 5,817,751, 5,990,112, 5,932,579 and 6,555,543, US Patent Application Publication Number 2004/0024180, PCT applications WO 96/00391 and WO 97/48685, and Smith et al., Bioorg. Med. Chem. Letters, 8, 2369- 2374 (1998).
For instance, diketopiperazines, such as DA-DKP, can be prepared by first synthesizing dipeptides. The dipeptides can be synthesized by methods well known in the
art using L-amino acids, D-amino acids or a combination of D- and L-amino acids. In some embodiments, solid-phase peptide synthetic methods are employed. Dipeptides are also available commercially from numerous sources, including DMI Synthesis Ltd., Cardiff, UK (custom synthesis), Sigma-Aldrich, St. Louis, MO (primarily custom synthesis), Phoenix Pharmaceuticals, Inc., Belmont, CA (custom synthesis), Fisher Scientific (custom synthesis) and Advanced ChemTech, Louisville, KY.
Once the dipeptide is synthesized or purchased, it is cyclized to form a diketopiperazine. This can be accomplished by a variety of techniques including, for example, the method provided in U.S. Patent Application Publication Number 2004/0024180, which describes a method of cyclizing dipeptides. Briefly, the dipeptide is heated in an organic solvent while removing water by distillation. In some embodiments, the organic solvent is a low-boiling azeotrope with water, such as acetonitrile, allyl alcohol, benzene, benzyl alcohol, n-butanol, 2-butanol, t-butanol, acetic acid butylester, carbon tetrachloride, chlorobenzene chloroform, cyclohexane, 1,2-dichlorethane, diethylacetal, dimethylacetal, acetic acid ethylester, heptane, methylisobutylketone, 3-pentanol, toluene and xylene. The temperature depends on the reaction speed at which the cyclization takes place and on the type of azeotroping agent used. In some embodiments, the reaction is carried out at 50-200°C, in some embodiments at 80-150°C. The pH range in which cyclization takes place can be 2-9, in some embodiments 3-7.
When one or both of the amino acids of the dipeptide has, or is derivatized to have, a carboxyl group on its side chain ( e.g ., aspartic acid or glutamic acid), the dipeptide is cyclized, for example as described in U.S. Patent No. 6,555,543. Briefly, the dipeptide, with the side-chain carboxyl still protected, is heated under neutral conditions. Typically, the dipeptide will be heated at from about 80°C to about 180°C, in some embodiments at about 120°C. The solvent will be a neutral solvent. For instance, the solvent may be an alcohol (such as butanol, methanol, ethanol, and higher alcohols, but not phenol) and an azeotropic co-solvent (such as toluene, benzene, or xylene). In some embodiments, the alcohol is butan-2-ol, and the azeotropic co-solvent is toluene. The heating is continued until the reaction is complete, and such times can be determined empirically. Typically, the dipeptide will be cyclized by refluxing it for about 8-24 hours, in some embodiments about 18 hours. Finally, the protecting group is removed from the diketopiperazine, preferably without the use of strong acids (mineral acids, such as sulfuric or hydrochloric acids), strong bases
(alkaline bases, such as potassium hydroxide or sodium hydroxide), and/or strong reducing agents ( e.g ., lithium aluminum hydride), in order to maintain the chirality of the final compound. In that regard, one or more of the compounds described herein contain one or more chiral centers and/or double bonds and, therefore, may exist as stereoisomers such as double-bond isomers (i.e., geometric isomers), enantiomers or diastereomers. Accordingly, the compounds described herein encompass all possible enantiomers and stereoisomers including the stereoisomerically pure form (e.g., geometrically pure, enantiomerically pure or diastereomerically pure) and enantiomeric and stereoisomeric mixtures. Enantiomeric and stereoisomeric mixtures can be resolved into their component enantiomers or stereoisomers using separation techniques or chiral synthesis techniques.
Dipeptides made on solid phase resins can be cyclized and released from the resin in one step (see, e.g, U.S. Patent No. 5,817,751). For instance, resin having an N-alkylated dipeptide attached can be suspended in toluene or toluene/ethanol in the presence of acetic acid (e.g., 1%) or triethylamine (e.g., 4%). Typically, basic cyclization conditions are utilized for their faster cyclization times.
Other methods of cyclizing dipeptides and of making diketopiperazines can be used in the preparation of diketopiperazines useful in the practice of the present disclosure. In addition, many diketopiperazines suitable for use in the present disclosure can be made as described below from proteins and peptides. Further, diketopiperazines for use in the practice of the present disclosure can be obtained commercially, for example from DMI Synthesis Ltd., Cardiff, UK (custom synthesis).
The DA-DKP compositions and/or products of the present disclosure can be prepared from solutions containing DA-DKP, including from commercially-available pharmaceutical compositions comprising albumin, such as human serum albumin, using methods such as ultrafiltration, chromatography, size-exclusion chromatography (e.g, Centricon filtration), affinity chromatography (e.g, using a column of beads having attached thereto an antibody or antibodies directed to the desired diketopiperazine(s) or an antibody or antibodies directed to the truncated protein or peptide), anion exchange or cation exchange, sucrose gradient centrifugation, salt precipitation, sonication, or other techniques that will remove some or all of the albumin in the solution. The resultant DA-DKP - containing compositions and/or products can be used and incorporated into pharmaceutical compositions as described above.
In some embodiments, the disclosed compositions and/or products can be prepared using an ultrafiltration separation method, whereby a human serum albumin composition can be passed over an ultrafiltration membrane having a molecular weight cut-off that retains the albumin while the DA-DKP passes into the resulting filtrate or fraction. This filtrate comprises components having molecular weights selected from less than about 50 kDa, less than about 40 kDa, less than 30 kDa, less than about 20 kDa, less than about 10 kDa, less than about 5 kDa, and less than about 3 kDa. In some embodiments, the filtrate is a LMWF comprising components having molecular weights less than about 5 kDa. This LMWF fraction or filtrate contains DA-DKP which is formed after the dipeptide aspartate-alanine is cleaved from albumin and subsequently cyclized into the diketopiperazine.
AMPION® (Ampio Pharmaceuticals, Inc., Englewood, CO USA) is a LMWF (<5 kDa fraction) of human serum albumin (HSA). AMPION® can be produced as described herein, for example by filtering commercially available HSA. Commercially available HSA is produced by fractionation of blood, for example by the Cohn process or its variations. AMPION® can be produced by filtering commercially available HSA, for example a 5% HSA solution, to remove components above 5 kDa.
Physiologically-acceptable salts of the DA-DKP of the present disclosure may also be used in the practice of the present disclosure. Physiologically-acceptable salts include conventional non-toxic salts, such as salts derived from inorganic acids (such as hydrochloric, hydrobromic, sulfuric, phosphoric, nitric, and the like), organic acids (such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, glutamic, aspartic, benzoic, salicylic, oxalic, ascorbic acid, and the like) or bases (such as the hydroxide, carbonate or bicarbonate of a pharmaceutically-acceptable metal cation or organic cations derived from N,N-dibenzylethylenediamine, D-glucosamine, or ethylenediamine). The salts are prepared in a conventional manner, for example by neutralizing the free base form of the compound with an acid.
Combination Therapy
In a second aspect provided by the present disclosure, methods of treating a viral respiratory disease are disclosed, the methods comprising administration of the disclosed compositions in combination with one or more additional therapeutics for the treatment of the viral respiratory disease. In various embodiments, the methods relate to the treatment of
a viral respiratory disease by administration of two or more therapeutics, each independently formulated to treat the viral respiratory disease. The two therapeutics can be administered concurrently or at different time points, via the same route or via different routes. In some embodiments, the two therapeutics are selected to administer complimentary, combination therapy to a subject suffering from a viral respiratory disease. Combination therapy is a treatment modality that combines two or more therapeutic agents. The amalgamation of therapeutic agents to treat viral respiratory diseases may enhance efficacy compared to the mono-therapy approach, as the therapeutic agents can be selected to target key pathways in a synergistic and/or additive manner. This approach has many benefits, including potentially reducing drug resistance, while simultaneously providing therapeutic benefits.
In some embodiments, the methods comprise administration of a first drug effective to treat a viral respiratory disease, for example COVID-19, and also administering a pharmaceutical composition provided by the present disclosure, comprising DA-DKP, to treat the same viral respiratory disease. In some embodiments, the methods comprise administration of a first drug before a hyperinflammatory stage of a viral respiratory disease and administering a pharmaceutical composition provided by the present disclosure, comprising DA-DKP, to treat a hyperinflammatory stage of the viral respiratory disease. The pharmaceutical composition comprising DA-DKP can be administered before, at, or after onset of the hyperinflammatory stage of the viral respiratory disease.
The first drug can be one or more antiviral, immune-modifying, anti-depressant, and/or corticosteroid drugs, including combinations thereof. In some embodiments, the first drug is selected from: Protease inhibitors: Paxlovid (PF-07321332) - a protease inhibitor antiviral therapy, nirmatrelvir (an orally active 3C-like protease inhibitor), ritonavir (protease and cytochrome P450-3A4 (CYP3A4) inhibitor); Nucleoside analogs (or prodrugs thereof): Molnupiravir - a prodrug of the synthetic nucleoside derivative N4- hydroxycytidine, Remdesivir - a prodrug of nucleoside analog GS-441524, Favipiravir - a prodrug of purine nucleic acid analog favipiravir-ribofuranosyl-5'-triphosphate; Immune- modifying antibodies: Tocilizumab - a monoclonal antibody against the interleukin-6 receptor, Sarilumab - a monoclonal antibody against the interleukin-6 receptor, Lenzilumab - monoclonal antibody against the cytokine granulocyte macrophage colony-stimulating factor (GM-CSF); Neutralizing antibodies: Casirivimab and imdevimab (REGN-COV) - monoclonal antibodies that bind to the spike protein of SARS-CoV-2, Bbamlanivimab and
etesevimab - monoclonal antibodies that bind to the spike protein of SARS-CoV-2, Sotrovimab - mAb that binds to the spike protein of SARS-CoV-2, Tixagevimab/cilgavimab (AZD7442) - mAb that binds to the spike protein of SARS-CoV- 2, Convalescent plasma; Other drugs: Baricitinib - a Janus kinase (JAK) inhibitor that reversibly inhibits Janus kinase 1 and Janus kinase 2, Tofacitinib - an inhibitor of the Janus kinase 1 (JAK1) and janus kinase 3 (JAK 3); Anticoagulation drugs: Fluvoxamine - an anti depressant selective serotonin reuptake inhibitor (SSRI), Recombinant ACE-2 - a decoy that is bound by SARS-CoV-2, EXO-CD24 - exosomes carrying CD24 (also known as signal transducer CD24, cluster of differentiation 24, or heat stable antigen CD24); Corticosteroids: Dexamethasone, Prednisone, Methylprednisolone, Hydrocortisone; and combinations of any of the foregoing.
EXAMPLES
AMPION® reduces CL075- and CL307 -induced CXCL10 release from PMA-differentiated THP-1 cells.
To determine if AMPION® influences endosomal single stranded ribonucleic acid (ssRNA) sensing pathways, CL075 and CL307 agonists were used to activate TLR7/8 or TLR7, respectively, in PMA-differentiated THP-1 cells. Increases in CXCL10 release indicate progression to hyperinflammation, and decreases indicate inhibition of progression to hyperinflammation.
AMPION® was manufactured by Ampio Pharmaceuticals, Inc. (Englewood, CO). General cell culture reagents were purchased from ThermoFisher Scientific (Waltham, MA) while X-Vivo 15 serum-free medium was obtained from Lonza (Basel, Switzerland). 0.9% (w/v) sodium chloride was obtained from KD Medical (Columbia, MD). GW9662 and CH223191 antagonists were purchased from MilliporeSigma (St. Louis, MO) with 100 mM stock solutions prepared in DMSO and stored at -80°C prior to use. CL075 and CL307 were purchased from Invivogen (San Diego, CA) and stock solutions prepared using supplied sterile, pyrogen-free water. ELISAs for IPIO/CXCLIO (catalog # KAC2361) were purchased from ThermoScientific. All other reagents were obtained from MilliporeSigma (St. Louis, MO) unless otherwise stated.
Cell Culture and Experimental Treatments
Human THP-1 monocytes (ATCC, Manassas, VA) were passaged in RPMI 1640 media supplemented with 10% fetal bovine serum (FBS) and 1% penicillin/ streptomycin
(Pen/ Strep) then differentiated using a final concentration of 50 ng/ml phorbol 12-myristate (PMA) in 96-well flat bottom plates seeded at 1 c 105 cells per well for 72 hours. Differentiation mediums were then aspirated and replenished as described below.
When CXCL10 release was evaluated 24 hours post-stimulation with CL075, dose- dependent release of chemokine was observed with significant (p-value < 0.05) reductions of 92 ± 3% and 78 ± 6% by AMPION® treatment at final agonist concentrations of 5 pg/ml and 2 pg/ml, respectively (Fig. 1A). Complete attenuation of CXCL10 release was also observed with AMPION® treatment at 0.5 pg/ml levels of CL075 but this may have been the result of release measurements at or near the detection limit of the ELISA. As for CL307- induced CXCL10 from PMA-differentiated THP-1 cells, muted chemokine release was observed at this timepoint only at the highest concentration used (1 pg/ml) with a trend towards inhibition (38 ± 29%; p-value = 0.5).
Different dose dynamics were observed when CXCL10 release was tested 72 hours post-stimulation (Fig. IB). Indicative of an asymptotic dose response, high levels of CL075- induced CXCL10 release was observed for all doses, with significant AMPION® inhibitions ranging from 62 ± 7% to 95 ± 3%. CXCL10 release in response to CL307 exposure at 72 hours was more vigorous and importantly, exhibited dose dependency. Drug-induced inhibitions of 84 ± 1% and 46 ± 23% were observed for 1 pg/ml and 0.1 pg/ml final concentrations of CL307, respectively, with the 1 pg/ml group exhibiting significance (p- value < 0.05).
Together, these data provide evidence that AMPION® modulates CXCL10 release resulting from the activation of TLR7/8 in macrophage-like THP-1 cells.
AMPION® reduces CL075- and CL307 -induced CXCL10 release from Peripheral Blood Mononuclear Cells (PBMC).
In a similar fashion, AMPION® was also found to inhibit CXCL10 release from TLR4-activated PBMC. To establish if AMPION® can influence ex vivo CL075- and CL307-induced CXCL10, these agonists were used to activate TLR7/8 in PBMC cocultures.
Both frozen and freshly isolated PBMC from consented donors were used in this investigation. When frozen stocks were utilized, cryopreserved vials obtained from Zen-Bio (Research Triangle Park, NC) were thawed using a Thawstar Automated Cell Thawing System (BioLife Solutions, Bothell, WA) and then transferred dropwise to RPMI 1640
medium containing 10% human AB serum, 1% penicillin-streptomycin (Pen/Strep), and 2 U/mL RNase-free DNase (ThermoScientific). Freshly isolated cells were isolated from sodium heparinized whole blood using Polymorphprep (Alere Technologies, Oslo, Norway) and washed with Dulbecco’s phosphate buffer saline. The resulting cell suspensions were then centrifuged at 1000 rpm for 10 minutes and working suspensions prepared at 2 x 106 cells/mL in X-Vivo 15 or RPMI 1640 supplemented with 20% FBS, 2% Pen/Strep, 1% sodium bicarbonate, 7.5% solution, 1% 100 mM sodium pyruvate, 1% 100X MEM non- essential amino acid solution, and 1% 200 mM L-glutamine.
For experimental treatments, 100 mΐ of PBMC suspensions were added to 96-well tissue culture plates or 100 mΐ of the 20% FBS RPMI medium described above was added to THP-1 plate wells. The solutions were then mixed with an equal volume of sterile 0.9% sodium chloride or AMPION® drug solutions and incubated at 37 °C and 5% CO2 for one hour. Stimulation was then achieved by adding CL075 or CL307 to the final concentrations indicated for an additional 24 to 72 hours before subsequent analysis. For relative potency investigation, 20 mM (10 mM final) concentration of antagonists were added to mediums prior to addition of saline or drug solutions serially diluted in saline. After the indicated stimulation periods described in these Examples, supernatants were collected for in-house CXCL10 ELISA measurements.
As observed in PMA-differentiated THP-1, CXCL10 release induced by CL075 from PBMC is observed 24 hours following exposure (Fig. 2A). However, an inverse relationship of CXCL10 release to CL075 concentration was observed using the donor cells represented in Fig. 2A, suggesting a biphasic response. AMPION® treatment resulted in significant inhibitions of 93 ± 10%, 55 ± 4%, and 65 ± 2% in CXCL10 release from 5 pg/ml, 2 pg/ml, and 0.5 pg/ml concentrations of agonist, respectively, when corrected for the background chemokine release observed for this donor. In contrast to THP-1, substantial CXCL10 release in response to a final concentration of 1.0 pg/ml CL307 was observed in PBMC cultures after 24 hours with a trend toward AMPION®-induced inhibition of 82 ± 33% (Fig. 2A). Measuring CXCL10 release from PBMC after 48 hours shows a similar asymptotic response compared to THP-1 cells with AMPION® inhibitions of 73 ± 2%, 68 ± 4%, and 76 ± 5% in CXCL10 release from 5 pg/ml, 2 pg/ml, and 0.5 pg/ml CL075, respectively (Fig. 2B). Significant inhibition of CXCL10 induced by 1.0 pg/ml CL307 was observed at this timepoint in the drug treatment group (81 ± 6%; p-value < 0.05).
These findings demonstrate AMPION® modulates TLR7/8 signaling in primary cells, such as monocytic and/or other myeloid cells.
Percent inhibition in CXCL10 release normalizes 24-hour CL075-induced PMA- differentiated THP-1 and PBMC data
Due to the magnitude and robustness of AMPION® inhibition in CXCL10 following 24-hour, 5 pg/ml CL075 activation, this treatment group was selected to determine variability and distributions in CXCL10 release. As shown in Fig. 3A, CL075-induced CXCL10 release from saline-treated PMA-differentiated THP-1 cells exhibits a wide, slightly negative distribution (Median = 4967 pg/ml, IQR = 4432, n = 8) with a distinct reduction seen in the AMPION® treatment group (Median = 276 pg/ml, IQR = 244, n = 8). CL075-induced CXCL10 release from saline-treated PBMC also exhibits a wide distribution with a strong positive skew (Median = 177 pg/ml, IQR = 756, n = 9 across a total of 7 donors) (Fig. 3B). Donor variability makes direct interpretation of raw release difficult, but a reduction in CXCL10 release was observed (Median = 121 pg/ml, IQR = 427, n = 9).
Fig. 3C demonstrates how normalization within each experiment to percent inhibition in CXCL10 release versus control corrected for observed background chemokine release and transformed the data to a more symmetrical distribution. Once drug response was calculated in this manner, AMPION® demonstrated an ability to inhibit CL075-induced CXCL10 release from PMA-differentiated THP-1 and PBMC by medians of 92% (IQR = 7, p-value < 0.05 versus hypothetical 0) and 58% (IQR = 34, p-value < 0.05 versus hypothetical 0) respectively.
Together, these data provide evidence that AMPION® modulates TLR7/8 signaling in both macrophage-like THP-1 cells and ex vivo PBMC cultures. Furthermore, the variability observed in overall CL075-induced CXCL10 release in PMA-differentiated THP-1 and PBMC models suggests that responses to TLR7/8 are highly dependent on cell- type and/or differentiation status. However, AMPION® inhibition of this pathway is universal, regardless of these dynamics.
PPARy and AhR antagonists reduce AMPION® drug potency as measured by CL075- induced CXCL10 release.
Inhibition of CXCL10 release from PMA-differentiated THP-1 cells was used to determine the involvement of various pathways. In the first step of this experimental
procedure, drug product was serially diluted with saline vehicle and then reductions in 24- hour CL075-induced CXCL10 release were evaluated following percent inhibition normalization. Regression analysis demonstrated that AMPION® exhibited a log-linear, dose-dependent reduction (R2 = 0.986) in chemokine release conducive for relative potency 5 (REP) calculation (Fig. 4). Next, the potency of AMPION® was established in the presence of antagonists for PPARy (GW9662, MilliporeSigma) or AhR (CH223191, Millipore Sigma) as compared to DMSO vehicle controls. Exposure of cells to 10 mM final concentrations of GW9662 or CH223191 resulted in shifts in the log-linear dose response of AMPION®- induced CXCL10 inhibition reflective of a loss in drug potency. REP calculated for 3 0 independent experiments demonstrated that antagonism of GW9662 and CH223191 resulted in reduced mean drug potency to 0.29 ± 0.06 and 0.40 ± 0.18, respectively (Table 1; data presented as mean relative potency (REP) ± STD and 95% Cl of three independent experiments versus DMSO controls). AMPION® thus activates PPARy and AhR transcription factors to inhibit the release of CXCL10 from PMA-differentiated THP-1 cells 5 stimulated with CL075.
ELISA array analysis demonstrates that AMPION * inhibits the release of a diverse set of pro-inflammatory cytokines and chemokines from CL075-activated, PMA-differentiated 0 THP-1 cells.
A multi-plex ELISA array was performed to assess the release of 48 cytokines, chemokines, and growth factors, to determine the overall immune response in PMA- differentiated THP-1 cells following CL075 activation and to identify additional drugs effects. The ELISA was Eve Technologies (Calgary, Alberta, Canada) human 5 cytokine/chemokine 48-plex Discovery Assay Array (catalog # HD48). Using this expanded format, a total of 26 inflammatory mediators were found to be significantly (p < 0.05) inhibited by AMPION®-treatment 24 hours following activation with 5 pg/ml CL075 (Table 2; data are presented as fold change ± STD or percent inhibition (n = 3, p-value = < 0.05 for
fold change versus theoretical fold change of 1)). Consistent with the previous findings in this investigation, CXCL10 proved to be the most highly downregulated (97 ± 2% inhibition) target on the array. Other prominent pro-inflammatory cytokines such as IL-6 (92 ± 9%), IL-12 (p40 = 91 ± 1%; p70 = 63 ± 7%), IL-Ib (70 ± 10%), and TNF-a (56 ± 5%) as well as chemokines including MCP (MCP-1 = 90 ± 1%; MCP-3 = 86 ± 5%), MIP (MIP- 1b = 70 ± 10%; MIR-1a = 68 ± 5%), CXCL9 (57 ± 11%) and RANTES (53 ± 1%) were also significantly suppressed. Of note, some reduction in type I and type II interferon was also observed but did not prove significant vs controls.
Table 2. Multiple cytokines and chemokines are significantly suppressed by AMPION® in CL075-stimulated, PMA-differentiated THP-1 cultures.
These quantified ELISA array results were then utilized to calculate biologically relevant cytokine ratios (Table 3; data presented as mean ratios ± STD, n 3, /i- value = < 0.05), which can be used to assess the immunological state of cells during inflammation. The ratio of IL-12 to IL-10, for example, has been established as a predictive marker of clinical course in multiple sclerosis as well as disease severity in viral infections. As shown in Table 3, treatment of CL075 -activated, PMA-differentiated THP-1 cells with AMP5A significantly reduced the ratio of IL-12p40/IL-10 released from 10.3 ± 3.5 seen in the saline control group to 1.3 ± 0.7 (p-value = 0.01). Similarly, IL-6/IL-10 ratios can be used to predict the prognosis of multiorgan dysfunction syndrome and mortality after trauma as well as the severity of disease in both pneumonia and COVID-19. As with IL-12/IL-10, AMP5A treatment significantly reduced IL-6/IL-10 ratio of these cultures (8.0 ± 4.1 for saline versus 0.6 ± 0.2; p-v alue = 0.04). These findings suggest that AMP5A is acting to biologically modulate the function of these cells in meaningful ways, as measured by phenotypic change, rather than simply inhibiting cytokine release.
In silico analysis of array cytokines and chemokines reveals that proteins inhibited by AMP ION® are associated with hyper -inflammatory disease states, immune cell maturation, and pro-inflammatory transcription factor activity.
Gene enrichment analysis was performed by querying Enrichr (Chen etal., Enrichr: interactive and collaborative HTML5 gene list enrichment analysis tool. BMC Bioinformatics. 2013; 14: 128.; Kuleshov MV., et al., Enrichr: a comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res. 2016;44:W90-7.) or Ingenuity Pathway Analysis (IP A) software using the corresponding gene symbols (Tables 4 and 5; data presented as tables of significantly enriched terms in Wikipathways and Jensens Disease libraries, ranked by enrichment score).
When sorted by enrichment score, the top term returned from the Wikipathways library of Enrichr was the ‘COVID-19 adverse outcome pathway’ (WP4891) followed by others such as ‘cytokine responses and inflammatory response’ (WP530) or ‘toll-like receptor signaling’ (WP75) as well as Tung fibrosis’ (WP3624) and ‘lymphoid development/polarization’ (WP3893 and WP4494). In addition, mining of the Jensen disease-associated databases predicts that these AMPION®-inhibited cytokines and chemokines are associated with lung disease, influenza, collagen disease, and arthritis.
Canonical pathway analysis performed in Ingenuity Pathway Analysis (IP A) showed that the cytokines and chemokines inhibited by AMPION® were predicted to significantly overlap with 6 pathways, and of these, ‘dendritic cell maturation’, crosstalk between dendritic cells and natural killer cells’, and ‘NF-kB signaling’ pathways were predicted to be significantly inhibited (black bars, Fig. 5) while IPA predicted activation of ‘liver X receptor/retinoid X receptor activity’ (grey bars, Fig. 5). The data demonstrate that
hyperinflammatory disease states, immune cell maturation, and proinflammatory transcription factor activity are suppressed with AMPION® treatment.
Data and Statistical Analysis
Statistical analysis was performed using the Real Statistics Resource Pack Excel Add-in (http://www.real-statistics.com/) unless otherwise stated. For representative ELISAs, two-tailed, two-sample unequal variance student tests were used to compare groups in Microsoft Excel (Microsoft Corporation, Redmond, WA). For combined inhibition of release analysis, two-tailed, one-sample t-tests (hypothetical value = 0; a = 0.05) were used to establish meaningful percent inhibition measurements from vehicle- controls. For cytokine arrays, two-tailed, one-sample t-tests were used to test for the significance of combined fold changes (hypothetical value = 1; a = 0.05). For potency assays, relative potency was calculated using 4P modeling with ANOVA pure separation and similarity of dose responses was established by f-tests for non-parallelism, non linearity, and significance of response in PLA 3.0 (Stegmann Systems GmbH, Raiffeisenstr, Germany). Box plots were generated, and descriptive statistics performed, using BoxPlotR (shiny.chemgrid.org/boxplotr/). In silico pathway analysis of differentially expressed genes was performed using Enrichr (maayanlab.cloud/Enrichr) or Ingenuity Pathway Analysis (IP A) software (Qiagen Digital Insights, Redwood City, CA). For IP A, calculated differentially abundant cytokines/chemokines were uploaded, and a ‘Core’ expression analysis based on log ratios for analytes with a p-value<0.05 was run using our ‘user dataset’ as reference. IPA calculated overlap p-values and z-scores as confidence metrics when predicting canonical pathway associations. For overlap p-values, we considered the - log(pvalue)>1.3 as significant. IPA calculated z-scores where z > 0 predicts activation, and z < 0 predicts inhibition; an absolute value of 2 was used as a significance cutoff.
Multi-modal COVID-19 treatment
A patient hospitalized due to COVID-19 receives standard of care therapy with the addition of AMPION® to prevent, reduce the severity or duration of, or treat a hyperinflammatory stage of COVID-19. The patient is administered remdesivir, remdesivir plus dexamethasone, or dexamethasone. The patient may also be administered one or more other antiviral drugs. The patient is administered AMPION® before, at, or after onset of the
hyperinflammatory stage. If the patient experiences rapidly increasing oxygen needs and systemic inflammation, baricitinab, tocilizumab, sarilumab, and/or another immunomodulatory drug may be administered. The antiviral drug or drugs inhibit the viral infection, and AMPION® treats the hyperinflammatory stage of COVID-19.
Claims
1. A method of treating one or more symptoms of a viral infection in a patient, comprising: administering a first drug to the patient prior to onset of a hyperinflammatory stage of the infection; and administering a pharmaceutical composition comprising DA-DKP to the patient before, at, or after the onset of the hyperinflammatory stage.
2. The method of claim 1, wherein the pharmaceutical composition is administered before the onset of the hyperinflammatory stage.
3. The method of claim 1, wherein the pharmaceutical composition is administered at the onset of the hyperinflammatory stage.
4. The method of claim 1, wherein the pharmaceutical composition is administered after the onset of the hyperinflammatory stage.
5. The method of any one of claims 1 - 4, wherein, upon administration of the pharmaceutical composition, the first drug and the pharmaceutical composition are co administered to the patient.
6. The method of any one of claims 1 - 5, wherein the first drug is an antiviral, an immune-modifying drug, an anti-depressant, a corticosteroid, or combinations thereof.
7. The method of any one of claims 1 - 5, wherein the first drug is selected from: a protease inhibitor selected from paxlovid, nirmatrelvir, and ritonavir; a nucleoside analog, or prodrug thereof, selected from molnupiravir, L - hydroxycytidine, remdesivir, favipiravir, and favipiravir-ribofuranosyl-5'-triphosphate; an immune-modifying antibody selected from tocilizumab, sarilumab, and lenzilumab; a neutralizing antibody selected from casirivimab, imdevimab, bamlanivimab, etesevimab, sotrovimab, tixagevimab/cilgavimab, and convalescent plasma; baricitinib or tofacitinib; an anti coagulation drug selected from fluvoxamine, recombinant ACE-2, and EXO-
CD24; a corticosteroid selected from dexamethasone, prednisone, methylprednisolone, and hydrocortisone; and
combinations of any of the foregoing.
8. The method of any one of claims 1 - 5, wherein the first drug is a protease inhibitor selected from paxlovid, nirmatrelvir, ritonavir, and combinations thereof.
9. The method of any one of claims 1 - 5, wherein the first drug is a nucleoside analog, or prodrug thereof, selected from molnupiravir, A -hy droxy cy ti dine, remdesivir, favipiravir, favipiravir-ribofuranosyl-5'-triphosphate, and combinations thereof.
10. The method of any one of claims 1 - 5, wherein the first drug is an immune- modifying antibody selected from tocilizumab, sarilumab, lenzilumab, and combinations thereof.
11. The method of any one of claims 1 -5, wherein the first drug is a neutralizing antibody selected from casirivimab, imdevimab, bamlanivimab, etesevimab, sotrovimab, tixagevimab/cilgavimab, convalescent plasma, and combinations thereof.
12. The method of any one of claims 1 - 5, wherein the first drug is selected from baricitinib, tofacitinib, and both baricitinib and tofacitinib.
13. The method of any one of claims 1 - 5, wherein the first drug is an anti coagulation drug selected from fluvoxamine, recombinant ACE-2, EXO-CD24, and combinations thereof.
14. The method of any one of claims 1 - 5, wherein the first drug is a corticosteroid selected from dexamethasone, prednisone, methylprednisolone, hydrocortisone, and combinations thereof.
15. The method of any one of claims 1 - 14, wherein the viral infection is caused by a respiratory virus.
16. The method of any one of claims 1 - 14, wherein the viral infection is caused by a virus selected from the group consisting of an influenza virus, a coronavirus, a rhinovirus, a respiratory syncytial virus, a parainfluenza virus, a human metapneumovirus, an adenovirus, and combinations thereof.
17. The method of any one of claims 1 - 14, wherein the viral infection is caused by a virus selected from the group consisting of an influenza A virus, an influenza B virus, Severe Acute Respiratory Distress Syndrome coronavirus (SARS-CoV), Middle East Respiratory Syndrome coronavirus (MERS-CoV), Severe Acute Respiratory Distress Syndrome coronavirus 2 (SARS-CoV-2), human adenovirus B21 infections (HAdV-B21), and combinations thereof.
18. The method of any one of claims 1-17, wherein the one or more symptoms are selected from the group consisting of acute respiratory distress syndrome (ARDS), acute lung injury (ALI), interstitial lung disease, pulmonary fibrosis, pneumonia, reactive airway disease syndrome, respiratory distress requiring supplemental oxygen, long COVID, and combinations thereof.
19. The method of any one of claims 5 - 18, wherein the co-administration results in an outcome selected from the group consisting of reduced ventilator time, reduced mortality, improvement in oxygenation parameters, reduced time to resolution of one or more respiratory symptoms, improved pulmonary function, and combinations thereof.
20. The method of any one of claims 5 - 19, wherein the co-administration results in the patient achieving improvement on the World Health Organization COVID- 19 ordinal scale of at least 4, at least 3, at least 2, or at least 1.
21. The method of any one of claims 1-20, wherein the patient has or had respiratory distress requiring supplemental oxygen caused by the viral infection.
22. The method of any one of claims 1-21, wherein the pharmaceutical composition is administered via inhalation.
23. The method of any one of claims 1-21, wherein the pharmaceutical composition is administered intravenously.
24. The method of any one of claims 1-23, wherein the pharmaceutical composition further comprises N-acetyl -tryptophan (NAT), caprylic acid, and/or caprylate.
25. A method of treating inflammation during a hyperinflammatory stage of a TLR7- and/or TLR8-mediated disease in a patient, comprising: administering a first drug to the patient prior to onset of a hyperinflammatory stage of the disease; and administering a pharmaceutical composition comprising DA-DKP to the patient before, at, or after the onset of the hyperinflammatory stage.
26. The method of claim 25, wherein the pharmaceutical composition is administered before the onset of the hyperinflammatory stage.
27. The method of claim 25, wherein the pharmaceutical composition is administered at the onset of the hyperinflammatory stage.
28. The method of claim 25, wherein the pharmaceutical composition is administered after the onset of the hyperinflammatory stage.
29. The method of any one of claims 25 - 28, wherein, upon administration of the pharmaceutical composition, the first drug and the pharmaceutical composition are co administered to the patient.
30. The method of any one of claims 25 - 29, wherein the TLR7- and/or TLR8- mediated disease is caused by a single-stranded RNA virus.
31. The method of any one of claims 25 - 30, wherein the TLR7- and/or TLR8- mediated disease is caused by a virus selected from the group consisting of Coronaviridae, SARS-CoV, MERS, SARS-CoV-2, torovirus, Hepeviridae, Hepatitis E, Caliciviridae, Norovirus, Sapporo virus, Togaviridae, Alphavirus viral diseases, Chikungunya, Eastern equine encephalomyelitis virus, Getah virus, Mayaro virus, Mucambo virus, O’nyong’nyong virus, Ross river virus, Barmah forest virus, Sagiyama virus, Semliki forest virus, Sindbis virus, Tonate virus, Venezuelan equine encephalomyelitis virus, Western equine encephalomyelitis virus and Rubivirus, Rubella virus, Flaviviridae, Hepacivirus, Hepatitis C, Flavivirus, Dengue virus type 1, Dengue virus type 2, Dengue virus type 3, Dengue virus type 4, Hepatitis G virus, Japanese B encephalitis virus, Murray Valley encephalitis virus, Rocio virus, Spondweni virus, St Louis encephalitis, Wesselsbron, West Nile virus, Yellow fever virus, Absettarov, Hanzalova, Hypr, Kumlinge, Kyasanur forest disease, Louping ill, Negishi, Omsk, Powassan, Langat, Russian spring summer encephalitis, Hepatitis G virus group, Hepatitis G virus, Picornaviridae, Enterovirus, Coxsackievirus, Echovirus, Poliovirus, Enterovirus 68-109, Rhinovirus A, Rhinovirus B, Hepatovirus, Hepatitis A, Astroviridae, Astrovirus species, Mononegavirales, Henipavirus, Hendra virus and Nipah virus, Rubulavirus, Mumps virus and Parainfluenza types 2, 4a and 4b, Morbillivirus, Measles virus, Avulavirus, Newcastle disease virus, Metapneumovirus, Pneumovirus, Respiratory syncytial virus, and Parainfluenza Type 1, Parainfluenza Type 2, Parainfluenza Type 3, Parainfluenza Type 4, Rhabdoviridae, Lyssavirus, Duvenhage and Rabies virus, Retroviridae, Human T-cell lymphotropic viruses, Lentivirus, Human Immunodeficiency virus, Arenaviridae, Arenavirus, lymphocytic choriomeningitis virus, Bunyaviridae, Bunyavirus, Bunyamwera virus, Hantavirus, Hantaan virus, Nairovirus, Nairobi sheep disease virus, and Phlebovirus, sandfly fever Sicilian virus, and Orthomyxoviridae, Influenza viruses, influenza virus A, influenza virus B, influenza virus C, influenza virus D, Thogoto-like viruses, Thogoto virus, and combinations thereof.
32. The method of any one of claims 25 - 30, wherein the TLR7- and/or TLR8- mediated disease is caused by a virus selected from the group consisting of a coronavirus, an influenza virus, respiratory syncytial virus (RSV), a parainfluenza virus, a human metapneumovirus, and combinations thereof.
33. The method of any one of claims 25 - 30, wherein the TLR7- and/or TLR8- mediated disease is caused by a virus selected from the group consisting of SARS- Coronavirus-2 (SARS-CoV-2), SARS-associated coronavirus (SARS-CoV), Middle East Respiratory Syndrome Coronavirus (MERS-CoV), and combinations thereof.
34. The method of any one of claims 25 - 30, wherein the TLR7- and/or TLR8- mediated disease is caused by SARS-Coronavirus-2 (SARS-CoV-2).
35. The method of any one of claims 25 - 30, wherein the TLR7- and/or TLR8- mediated disease is caused by a virus selected from the group consisting of an influenza A virus, an influenza B virus, and both an influenza A virus and an influenza B virus.
36. The method of any one of claims 25 - 29, wherein the TLR7- and/or TLR8- mediated disease is selected from the group consisting of acute kidney injury, glomerulonephritis, lupus, lupus nephritis, and combinations thereof.
37. The method of any one of claims 25 - 36, wherein the first drug is selected from: a protease inhibitor selected from paxlovid, nirmatrelvir, and ritonavir; a nucleoside analog, or prodrug thereof, selected from molnupiravir, N4- hydroxycytidine, remdesivir, favipiravir, and favipiravir-ribofuranosyl-5'-triphosphate; an immune-modifying antibody selected from tocilizumab, sarilumab, and lenzilumab; a neutralizing antibody selected from casirivimab, imdevimab, bamlanivimab, etesevimab, sotrovimab, tixagevimab/cilgavimab, and convalescent plasma; baricitinib or tofacitinib; an anti coagulation drug selected from fluvoxamine, recombinant ACE-2, and EXO-
CD24; a corticosteroid selected from dexamethasone, prednisone, methylprednisolone, and hydrocortisone; and combinations of any of the foregoing.
38. The method of any one of claims 25 - 36, wherein the first drug is a protease inhibitor selected from paxlovid, nirmatrelvir, ritonavir, and combinations thereof.
39. The method of any one of claims 25 - 36, wherein the first drug is a nucleoside analog, or prodrug thereof, selected from molnupiravir, A -hydroxycytidine, remdesivir, favipiravir, favipiravir-ribofuranosyl-5'-triphosphate, and combinations thereof.
40. The method of any one of claims 25 - 36, wherein the first drug is an immune-modifying antibody selected from tocilizumab, sarilumab, lenzilumab, and combinations thereof.
41. The method of any one of claims 25 - 36, wherein the first drug is a neutralizing antibody selected from casirivimab, imdevimab, bamlanivimab, etesevimab, sotrovimab, tixagevimab/cilgavimab, convalescent plasma, and combinations thereof.
42. The method of any one of claims 25 - 36, wherein the first drug is selected from baricitinib, tofacitinib, and both baricitinib and tofacitinib.
43. The method of any one of claims 25 - 36, wherein the first drug is an anti coagulation drug selected from fluvoxamine, recombinant ACE-2, EXO-CD24, and combinations thereof.
44. The method of any one of claims 25 - 36, wherein the first drug is a corticosteroid selected from dexamethasone, prednisone, methylprednisolone, hydrocortisone, and combinations thereof.
45. The method of any one of claims 30 - 35 or 37 - 44, wherein the patient has or had respiratory distress requiring supplemental oxygen caused by the viral infection.
46. The method of any one of claims 25 - 45, wherein the pharmaceutical composition is administered via inhalation.
47. The method of any one of claims 25 - 46, wherein the pharmaceutical composition is administered intravenously.
48. The method of any one of claims 25 - 47, wherein the pharmaceutical composition further comprises N-acetyl -tryptophan (NAT), caprylic acid, and/or caprylate.
49. A method of treating or preventing one or more symptoms of a TLR7- and/or TLR8-mediated disease in a patient, comprising administering to the patient a pharmaceutical composition comprising DA-DKP.
50. A method of treating or preventing inflammation associated with a TLR7- and/or TLR8-mediated disease in a patient, comprising administering to the patient a pharmaceutical composition comprising DA-DKP.
51. The method of claim 49 or claim 50, wherein the TLR7- and/or TLR8- mediated disease is caused by a single-stranded RNA virus.
52. The method of any one of claims 49 - 51, wherein the TLR7- and/or TLR8- mediated disease is caused by a virus selected from the group consisting of Coronaviridae, SARS-CoV, MERS, SARS-CoV-2, torovirus, Hepeviridae, Hepatitis E, Caliciviridae, Norovirus, Sapporo virus, Togaviridae, Alphavirus viral diseases, Chikungunya, Eastern equine encephalomyelitis virus, Getah virus, Mayaro virus, Mucambo virus, O’nyong’nyong virus, Ross river virus, Barmah forest virus, Sagiyama virus, Semliki forest virus, Sindbis virus, Tonate virus, Venezuelan equine encephalomyelitis virus, Western equine encephalomyelitis virus and Rubivirus, Rubella virus, Flaviviridae, Hepacivirus, Hepatitis C, Flavivirus, Dengue virus type 1, Dengue virus type 2, Dengue virus type 3, Dengue virus type 4, Hepatitis G virus, Japanese B encephalitis virus, Murray Valley encephalitis virus, Rocio virus, Spondweni virus, St Louis encephalitis, Wesselsbron, West Nile virus, Yellow fever virus, Absettarov, Hanzalova, Hypr, Kumlinge, Kyasanur forest disease, Louping ill, Negishi, Omsk, Powassan, Langat, Russian spring summer encephalitis, Hepatitis G virus group, Hepatitis G virus, Picornaviridae, Enterovirus, Coxsackievirus, Echovirus, Poliovirus, Enterovirus 68-109, Rhinovirus A, Rhinovirus B, Hepatovirus, Hepatitis A, Astroviridae, Astrovirus species, Mononegavirales, Henipavirus, Hendra virus and Nipah virus, Rubulavirus, Mumps virus and Parainfluenza types 2, 4a and 4b, Morbillivirus, Measles virus, Avulavirus, Newcastle disease virus, Metapneumovirus, Pneumovirus, Respiratory syncytial virus, and Parainfluenza Type 1, Parainfluenza Type 2, Parainfluenza Type 3, Parainfluenza Type 4, Rhabdoviridae, Lyssavirus, Duvenhage and Rabies virus, Retroviridae, Human T-cell lymphotropic viruses, Lentivirus, Human Immunodeficiency virus, Arenaviridae, Arenavirus, lymphocytic choriomeningitis virus, Bunyaviridae, Bunyavirus, Bunyamwera virus, Hantavirus, Hantaan virus, Nairovirus, Nairobi sheep disease virus, and Phlebovirus, sandfly fever Sicilian virus, and Orthomyxoviridae, Influenza viruses, influenza virus A, influenza virus B, influenza virus C, influenza virus D, Thogoto-like viruses, Thogoto virus, and combinations thereof.
53. The method of any one of claims 49 - 51, wherein the TLR7- and/or TLR8- mediated disease is caused by a virus selected from group consisting of a coronavirus, an influenza virus, respiratory syncytial virus (RSV), a parainfluenza virus, a human metapneumovirus, and combinations thereof.
54. The method of any one of claims 49 - 51, wherein the TLR7- and/or TLR8- mediated disease is caused by a virus selected from the group consisting of SARS- Coronavirus-2 (SARS-CoV-2), SARS-associated coronavirus (SARS-CoV), Middle East Respiratory Syndrome Coronavirus (MERS-CoV), and combinations thereof.
55. The method of any one of claims 49 - 51, wherein the TLR7- and/or TLR8- mediated disease is caused by SARS-Coronavirus-2 (SARS-CoV-2).
56. The method of any one of claims 49 - 51, wherein the TLR7- and/or TLR8- mediated disease is caused by a virus selected from the group consisting of an influenza A virus, an influenza B virus, and both an influenza A virus and an influenza B virus.
57. The method of any one of claims 49 - 56, wherein the patient has or is at risk of developing inflammation of a tissue selected from the group consisting of lung, brain, heart, kidney, blood vessel, skin, and nerve.
58. The method of claim 57, wherein the tissue is selected from the group consisting of lung and kidney.
59. The method of any one of claims 49 - 58, wherein the patient has or is at risk of developing a symptom selected from the group consisting of acute respiratory distress syndrome (ARDS), acute lung injury (ALI), interstitial lung disease, pulmonary fibrosis, pneumonia, reactive airway disease syndrome, and combinations thereof.
60. The method of any one of claims 49 - 59, wherein the patient has or is at risk of developing a symptom selected from the group consisting of fatigue, shortness of breath or difficulty breathing, low exercise tolerance, low blood oxygen saturation, cough, sore throat, stuffy or runny nose, joint pain, chest pain, tightness or discomfort, muscle pain, muscle weakness, fever, heart palpitations, difficulty thinking and/or concentrating, depression, and combinations thereof.
61. The method of claim 60, wherein the patient has experienced the symptom at least four weeks, at least one month, at least two months, or at least three months.
62. The method of any one of claims 49 - 61, wherein the administering results in an outcome selected from the group consisting of reduced ventilator time, reduced
mortality, improvement in oxygenation parameters, reduced time to resolution of one or more respiratory symptoms, improved pulmonary function, and combinations thereof.
63. The method of any one of claims 49 - 62, wherein, after the administration, the patient achieves improvement on the World Health Organization COVID-19 ordinal scale of at least 4, at least 3, at least 2, or at least 1.
64. The method of any one of claims 49 - 63, wherein the patient has respiratory distress.
65. The method of claim 64, wherein the patient requires supplemental oxygen.
66. The method of any one of claims 49 - 65, wherein the composition is administered by inhalation.
67. The method of claim 66, wherein the composition is administered by a nebulizer.
68. The method of claim 66 or claim 67, wherein 8 ml of the composition is administered to the patient.
69. The method of any one of claims 66 - 68, wherein the composition is administered quater in die.
70. The method of any one of claims 49 - 65, wherein the composition is administered intravenously.
71. The method of claim 70, wherein 250 cc of the composition is administered to the patient.
72. The method of claim 70 or claim 71, wherein the composition is administered bis in die.
73. The method of any one of claims 49 - 72, wherein the composition further comprises N-acetyl-tryptophan (NAT), caprylic acid, caprylate or combinations thereof.
74. The method of any one of claims 49 - 73, wherein the pharmaceutical composition is prepared by removing albumin from a solution of a human serum albumin composition.
75. The method of claim 74, wherein the albumin is removed by ultrafiltration, sucrose gradient centrifugation, chromatography, salt precipitation, sonication, or combinations thereof.
76. The method of claim 74 or claim 75, wherein the albumin is removed by passing the human serum albumin composition over an ultrafiltration membrane with a
molecular weight cut off that retains the albumin, wherein the resulting filtrate comprises DA-DKP.
77. The method of claim 76, wherein the ultrafiltration membrane has a molecular weight cutoff of less than 50 kDa, less than 40 kDa, less than 30 kDa, less than 20 kDa, less than 10 kDa, less than 5 kDa or less than 3 kDa.
78. The method of any one of claims 49, 50 or 57 - 77, wherein the TLR7- and/or TLR8-mediated disease is selected from the group consisting of lupus, lupus nephritis, and combinations thereof.
Applications Claiming Priority (4)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US202163169690P | 2021-04-01 | 2021-04-01 | |
US63/169,690 | 2021-04-01 | ||
US202263311755P | 2022-02-18 | 2022-02-18 | |
US63/311,755 | 2022-02-18 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2022212914A1 true WO2022212914A1 (en) | 2022-10-06 |
Family
ID=83456855
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2022/023162 WO2022212914A1 (en) | 2021-04-01 | 2022-04-01 | Methods of treating viral diseases |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2022212914A1 (en) |
Cited By (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN117554628A (en) * | 2024-01-11 | 2024-02-13 | 北京大学人民医院 | Inflammatory factor composition, model and kit for early warning of MODS |
CN117838697A (en) * | 2024-01-09 | 2024-04-09 | 暨南大学 | Application of baratinib in preparation of medicine for treating influenza virus infection |
CN117838696A (en) * | 2024-01-09 | 2024-04-09 | 暨南大学 | Application of ruxotinib in preparation of medicine for treating influenza virus infection |
WO2024081269A1 (en) * | 2022-10-10 | 2024-04-18 | Dixit Rohan | Therapeutic combinations and methods to treat long covid |
WO2024165081A1 (en) * | 2023-02-08 | 2024-08-15 | 南通市海门品尚医药科技有限公司 | Liquid preparation containing nirmatrelvir and ritonavir, and preparation method therefor and use thereof |
Citations (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20130345238A1 (en) * | 2000-08-04 | 2013-12-26 | Dmi Acquisition Corp. | Method of using diketopiperazines and composition containing them |
WO2018044724A1 (en) * | 2016-08-31 | 2018-03-08 | Ampio Pharmaceuticals, Inc. | Treatment of disease with n-acetyl kynurenine |
-
2022
- 2022-04-01 WO PCT/US2022/023162 patent/WO2022212914A1/en active Application Filing
Patent Citations (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20130345238A1 (en) * | 2000-08-04 | 2013-12-26 | Dmi Acquisition Corp. | Method of using diketopiperazines and composition containing them |
WO2018044724A1 (en) * | 2016-08-31 | 2018-03-08 | Ampio Pharmaceuticals, Inc. | Treatment of disease with n-acetyl kynurenine |
Non-Patent Citations (1)
Title |
---|
GARCIA CRISTIANA C., RUSSO REMO C., GUABIRABA RODRIGO, FAGUNDES CAIO T., POLIDORO RAFAEL B., TAVARES LUCIANA P., SALGADO ANA PAULA: "Platelet-Activating Factor Receptor Plays a Role in Lung Injury and Death Caused by Influenza A in Mice", PLOS PATHOGENS, vol. 6, no. 11, 4 November 2010 (2010-11-04), pages e1001171, XP055976197, DOI: 10.1371/journal.ppat.1001171 * |
Cited By (6)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2024081269A1 (en) * | 2022-10-10 | 2024-04-18 | Dixit Rohan | Therapeutic combinations and methods to treat long covid |
WO2024165081A1 (en) * | 2023-02-08 | 2024-08-15 | 南通市海门品尚医药科技有限公司 | Liquid preparation containing nirmatrelvir and ritonavir, and preparation method therefor and use thereof |
CN117838697A (en) * | 2024-01-09 | 2024-04-09 | 暨南大学 | Application of baratinib in preparation of medicine for treating influenza virus infection |
CN117838696A (en) * | 2024-01-09 | 2024-04-09 | 暨南大学 | Application of ruxotinib in preparation of medicine for treating influenza virus infection |
CN117554628A (en) * | 2024-01-11 | 2024-02-13 | 北京大学人民医院 | Inflammatory factor composition, model and kit for early warning of MODS |
CN117554628B (en) * | 2024-01-11 | 2024-03-12 | 北京大学人民医院 | Inflammatory factor composition, model and kit for early warning of MODS |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
WO2022212914A1 (en) | Methods of treating viral diseases | |
Najafloo et al. | Mechanism of anosmia caused by symptoms of COVID-19 and emerging treatments | |
Ashour et al. | A systematic review of the global intervention for SARS-CoV-2 combating: from drugs repurposing to molnupiravir approval | |
RU2524304C2 (en) | Application of acetylsalicylic acid salt for treatment of viral infections | |
Rohilla | Designing therapeutic strategies to combat severe acute respiratory syndrome coronavirus‐2 disease: COVID‐19 | |
Garcia-del-Barco et al. | Revisiting pleiotropic effects of type I interferons: rationale for its prophylactic and therapeutic use against SARS-CoV-2 | |
KR20230022164A (en) | How Bardoxolone Methyl or Its Analogs Are Used to Treat COVID-19 | |
Chen et al. | Sertraline is an effective SARS-CoV-2 entry inhibitor targeting the spike protein | |
CN115996741A (en) | CXCR4 inhibitors for the treatment of acute respiratory distress syndrome and viral infections | |
KR20220150348A (en) | PLD for use in combination in the treatment of coronavirus | |
Malkan et al. | Hematological aspects of the COVID-19 syndrome. | |
US20230149515A1 (en) | Inhibition of sars-cov-2 viral entry through oral administration of lactoferrin and uses thereof | |
JP2023553610A (en) | Interfering RNAS targeting severe acute respiratory syndrome-associated coronaviruses and their use to treat COVID-19 | |
KR20230018474A (en) | Formulations and methods for treating acute respiratory distress syndrome, asthma, or allergic rhinitis | |
Cuevas et al. | Fibroblast growth factor: A target for Covid-19 infection | |
JP2024517334A (en) | Pharmaceutical compositions and methods for the treatment of acute respiratory distress syndrome (ARDS) in coronavirus disease (COVID-19) | |
US20230233488A1 (en) | Novel use of a modulator of glucosylceramide degradation for viral infections | |
US10677782B2 (en) | Methods of determining interferon having direct inhibitory effects on tumors and uses thereof | |
Tufan et al. | Immune dysfunction in COVID-19 and judicious use of antirheumatic drugs for thetreatment of hyperinflammation | |
JPWO2011142484A1 (en) | Viral infection drug containing polyalkyleneimine | |
Kumar | A review on pharmacokinetics, pharmacodynamics and clinical aspects of remdesivir and favipiravir for the treatment of coronavirus disease | |
US10877024B2 (en) | Methods of determining interferon having direct inhibitory effects on tumors and uses thereof | |
US20220304947A1 (en) | Compositions and methods for inhibiting proteolytic activation of viruses | |
Parveen et al. | Investigational Drugs for the COVID 19 Pandemic–A Concise Review | |
Ismael | Article Review: Toll-like Receptors and COVID-19 |
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: 22782338 Country of ref document: EP Kind code of ref document: A1 |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
122 | Ep: pct application non-entry in european phase |
Ref document number: 22782338 Country of ref document: EP Kind code of ref document: A1 |