WO2024186623A1 - Methods of making dried pharmaceutical compositions - Google Patents
Methods of making dried pharmaceutical compositions Download PDFInfo
- Publication number
- WO2024186623A1 WO2024186623A1 PCT/US2024/018048 US2024018048W WO2024186623A1 WO 2024186623 A1 WO2024186623 A1 WO 2024186623A1 US 2024018048 W US2024018048 W US 2024018048W WO 2024186623 A1 WO2024186623 A1 WO 2024186623A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- lipid
- lipid vesicle
- vesicle particles
- mixture
- drying
- Prior art date
Links
- 238000000034 method Methods 0.000 title claims abstract description 280
- 239000008194 pharmaceutical composition Substances 0.000 title claims abstract description 59
- 150000002632 lipids Chemical class 0.000 claims abstract description 391
- 239000003814 drug Substances 0.000 claims abstract description 101
- 229940124597 therapeutic agent Drugs 0.000 claims abstract description 96
- 108091033319 polynucleotide Proteins 0.000 claims abstract description 88
- 102000040430 polynucleotide Human genes 0.000 claims abstract description 88
- 239000002157 polynucleotide Substances 0.000 claims abstract description 88
- 239000002671 adjuvant Substances 0.000 claims abstract description 83
- 239000002245 particle Substances 0.000 claims description 256
- 239000000427 antigen Substances 0.000 claims description 160
- 108091007433 antigens Proteins 0.000 claims description 160
- 102000036639 antigens Human genes 0.000 claims description 160
- 108090000765 processed proteins & peptides Proteins 0.000 claims description 148
- 238000002360 preparation method Methods 0.000 claims description 118
- 239000000203 mixture Substances 0.000 claims description 109
- 239000012528 membrane Substances 0.000 claims description 96
- 239000004417 polycarbonate Substances 0.000 claims description 65
- 229920000515 polycarbonate Polymers 0.000 claims description 65
- HVYWMOMLDIMFJA-DPAQBDIFSA-N cholesterol Chemical compound C1C=C2C[C@@H](O)CC[C@]2(C)[C@@H]2[C@@H]1[C@@H]1CC[C@H]([C@H](C)CCCC(C)C)[C@@]1(C)CC2 HVYWMOMLDIMFJA-DPAQBDIFSA-N 0.000 claims description 62
- IPCSVZSSVZVIGE-UHFFFAOYSA-N hexadecanoic acid Chemical compound CCCCCCCCCCCCCCCC(O)=O IPCSVZSSVZVIGE-UHFFFAOYSA-N 0.000 claims description 50
- 206010028980 Neoplasm Diseases 0.000 claims description 46
- 238000004108 freeze drying Methods 0.000 claims description 42
- 238000001035 drying Methods 0.000 claims description 39
- 230000028993 immune response Effects 0.000 claims description 39
- 102000004196 processed proteins & peptides Human genes 0.000 claims description 37
- 238000002156 mixing Methods 0.000 claims description 35
- SNKAWJBJQDLSFF-NVKMUCNASA-N 1,2-dioleoyl-sn-glycero-3-phosphocholine Chemical compound CCCCCCCC\C=C/CCCCCCCC(=O)OC[C@H](COP([O-])(=O)OCC[N+](C)(C)C)OC(=O)CCCCCCC\C=C/CCCCCCCC SNKAWJBJQDLSFF-NVKMUCNASA-N 0.000 claims description 34
- 201000011510 cancer Diseases 0.000 claims description 33
- 238000001125 extrusion Methods 0.000 claims description 32
- 235000012000 cholesterol Nutrition 0.000 claims description 31
- 238000004513 sizing Methods 0.000 claims description 31
- 239000012051 hydrophobic carrier Substances 0.000 claims description 30
- VMHLLURERBWHNL-UHFFFAOYSA-M Sodium acetate Chemical compound [Na+].CC([O-])=O VMHLLURERBWHNL-UHFFFAOYSA-M 0.000 claims description 29
- 239000002502 liposome Substances 0.000 claims description 29
- 239000001632 sodium acetate Substances 0.000 claims description 29
- 235000017281 sodium acetate Nutrition 0.000 claims description 29
- 239000012634 fragment Substances 0.000 claims description 28
- 239000001488 sodium phosphate Substances 0.000 claims description 28
- 229910000162 sodium phosphate Inorganic materials 0.000 claims description 28
- RYFMWSXOAZQYPI-UHFFFAOYSA-K trisodium phosphate Chemical compound [Na+].[Na+].[Na+].[O-]P([O-])([O-])=O RYFMWSXOAZQYPI-UHFFFAOYSA-K 0.000 claims description 28
- 108010002687 Survivin Proteins 0.000 claims description 27
- 150000003904 phospholipids Chemical class 0.000 claims description 27
- 239000000243 solution Substances 0.000 claims description 26
- 239000003795 chemical substances by application Substances 0.000 claims description 25
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 claims description 23
- 229920001184 polypeptide Polymers 0.000 claims description 22
- 235000021314 Palmitic acid Nutrition 0.000 claims description 21
- WQEPLUUGTLDZJY-UHFFFAOYSA-N n-Pentadecanoic acid Natural products CCCCCCCCCCCCCCC(O)=O WQEPLUUGTLDZJY-UHFFFAOYSA-N 0.000 claims description 20
- 230000002519 immonomodulatory effect Effects 0.000 claims description 18
- 239000003921 oil Substances 0.000 claims description 18
- 235000019198 oils Nutrition 0.000 claims description 18
- 229940126586 small molecule drug Drugs 0.000 claims description 18
- 241000725643 Respiratory syncytial virus Species 0.000 claims description 16
- 241000701806 Human papillomavirus Species 0.000 claims description 15
- 102000000763 Survivin Human genes 0.000 claims description 15
- 239000003960 organic solvent Substances 0.000 claims description 15
- 150000001413 amino acids Chemical class 0.000 claims description 14
- 238000011146 sterile filtration Methods 0.000 claims description 14
- 239000007864 aqueous solution Substances 0.000 claims description 13
- 125000003275 alpha amino acid group Chemical group 0.000 claims description 12
- 230000002452 interceptive effect Effects 0.000 claims description 12
- 238000011282 treatment Methods 0.000 claims description 12
- 241000193738 Bacillus anthracis Species 0.000 claims description 11
- 208000015181 infectious disease Diseases 0.000 claims description 11
- 238000000265 homogenisation Methods 0.000 claims description 10
- 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 claims description 9
- 108010028921 Lipopeptides Proteins 0.000 claims description 9
- 230000001939 inductive effect Effects 0.000 claims description 9
- 239000000787 lecithin Substances 0.000 claims description 9
- 229940067606 lecithin Drugs 0.000 claims description 9
- 235000010445 lecithin Nutrition 0.000 claims description 9
- 235000010446 mineral oil Nutrition 0.000 claims description 9
- 239000002480 mineral oil Substances 0.000 claims description 9
- 241000725303 Human immunodeficiency virus Species 0.000 claims description 8
- DKGAVHZHDRPRBM-UHFFFAOYSA-N Tert-Butanol Chemical compound CC(C)(C)O DKGAVHZHDRPRBM-UHFFFAOYSA-N 0.000 claims description 8
- 208000035473 Communicable disease Diseases 0.000 claims description 7
- 239000007921 spray Substances 0.000 claims description 7
- 238000001694 spray drying Methods 0.000 claims description 7
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N Dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 claims description 6
- 239000002243 precursor Substances 0.000 claims description 6
- 229940065181 bacillus anthracis Drugs 0.000 claims description 5
- 238000011068 loading method Methods 0.000 claims description 5
- 230000000717 retained effect Effects 0.000 claims description 5
- 108010084333 N-palmitoyl-S-(2,3-bis(palmitoyloxy)propyl)cysteinyl-seryl-lysyl-lysyl-lysyl-lysine Proteins 0.000 claims description 4
- 241000224016 Plasmodium Species 0.000 claims description 4
- 230000015556 catabolic process Effects 0.000 claims description 4
- 238000006731 degradation reaction Methods 0.000 claims description 4
- 238000004128 high performance liquid chromatography Methods 0.000 claims description 4
- 230000005847 immunogenicity Effects 0.000 claims description 4
- BVAUMRCGVHUWOZ-ZETCQYMHSA-N (2s)-2-(cyclohexylazaniumyl)propanoate Chemical group OC(=O)[C@H](C)NC1CCCCC1 BVAUMRCGVHUWOZ-ZETCQYMHSA-N 0.000 claims description 3
- 229920001519 homopolymer Polymers 0.000 claims description 3
- COLNVLDHVKWLRT-UHFFFAOYSA-N phenylalanine Natural products OC(=O)C(N)CC1=CC=CC=C1 COLNVLDHVKWLRT-UHFFFAOYSA-N 0.000 claims description 3
- COLNVLDHVKWLRT-QMMMGPOBSA-N phenylalanine group Chemical group N[C@@H](CC1=CC=CC=C1)C(=O)O COLNVLDHVKWLRT-QMMMGPOBSA-N 0.000 claims description 3
- 238000000527 sonication Methods 0.000 claims description 3
- AKIIJJAKZRNOLW-HTIIIDOHSA-N (2r)-2-[di(hexadecanoyl)amino]-3-(2,3-dihydroxypropylsulfanyl)propanoic acid Chemical compound CCCCCCCCCCCCCCCC(=O)N([C@@H](CSCC(O)CO)C(O)=O)C(=O)CCCCCCCCCCCCCCC AKIIJJAKZRNOLW-HTIIIDOHSA-N 0.000 claims description 2
- PZFZLRNAOHUQPH-DJBVYZKNSA-N (2r)-3-[(2r)-2,3-di(hexadecanoyloxy)propyl]sulfanyl-2-(hexadecanoylamino)propanoic acid Chemical compound CCCCCCCCCCCCCCCC(=O)N[C@H](C(O)=O)CSC[C@H](OC(=O)CCCCCCCCCCCCCCC)COC(=O)CCCCCCCCCCCCCCC PZFZLRNAOHUQPH-DJBVYZKNSA-N 0.000 claims description 2
- 108010038122 S-(2,3-bis(palmitoyloxy)propyl)cysteine Proteins 0.000 claims description 2
- 229920000140 heteropolymer Polymers 0.000 claims description 2
- 235000019488 nut oil Nutrition 0.000 claims description 2
- 239000010466 nut oil Substances 0.000 claims description 2
- 229940049964 oleate Drugs 0.000 claims description 2
- 235000015112 vegetable and seed oil Nutrition 0.000 claims description 2
- 239000008158 vegetable oil Substances 0.000 claims description 2
- HEMHJVSKTPXQMS-UHFFFAOYSA-M Sodium hydroxide Chemical compound [OH-].[Na+] HEMHJVSKTPXQMS-UHFFFAOYSA-M 0.000 claims 12
- QTBSBXVTEAMEQO-UHFFFAOYSA-N Acetic acid Chemical compound CC(O)=O QTBSBXVTEAMEQO-UHFFFAOYSA-N 0.000 claims 6
- 238000007789 sealing Methods 0.000 claims 4
- 230000003381 solubilizing effect Effects 0.000 claims 2
- ZQPPMHVWECSIRJ-KTKRTIGZSA-N oleic acid Chemical compound CCCCCCCC\C=C/CCCCCCCC(O)=O ZQPPMHVWECSIRJ-KTKRTIGZSA-N 0.000 claims 1
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 28
- 108090000623 proteins and genes Proteins 0.000 description 27
- 150000001875 compounds Chemical class 0.000 description 22
- 241000282414 Homo sapiens Species 0.000 description 21
- 210000004027 cell Anatomy 0.000 description 21
- -1 diglycerides Chemical class 0.000 description 21
- 230000002209 hydrophobic effect Effects 0.000 description 21
- 201000010099 disease Diseases 0.000 description 20
- 102000004169 proteins and genes Human genes 0.000 description 20
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 19
- 235000018102 proteins Nutrition 0.000 description 18
- QFJCIRLUMZQUOT-HPLJOQBZSA-N sirolimus Chemical compound C1C[C@@H](O)[C@H](OC)C[C@@H]1C[C@@H](C)[C@H]1OC(=O)[C@@H]2CCCCN2C(=O)C(=O)[C@](O)(O2)[C@H](C)CC[C@H]2C[C@H](OC)/C(C)=C/C=C/C=C/[C@@H](C)C[C@@H](C)C(=O)[C@H](OC)[C@H](O)/C(C)=C/[C@@H](C)C(=O)C1 QFJCIRLUMZQUOT-HPLJOQBZSA-N 0.000 description 16
- 239000000126 substance Substances 0.000 description 15
- 235000001014 amino acid Nutrition 0.000 description 14
- 210000003719 b-lymphocyte Anatomy 0.000 description 13
- 210000001744 T-lymphocyte Anatomy 0.000 description 12
- 241000700605 Viruses Species 0.000 description 12
- 229960005486 vaccine Drugs 0.000 description 12
- 108020004414 DNA Proteins 0.000 description 11
- 125000003473 lipid group Chemical group 0.000 description 11
- 244000052769 pathogen Species 0.000 description 11
- 239000012071 phase Substances 0.000 description 11
- 108060003951 Immunoglobulin Proteins 0.000 description 10
- 102000018358 immunoglobulin Human genes 0.000 description 10
- 239000010410 layer Substances 0.000 description 10
- 230000001717 pathogenic effect Effects 0.000 description 10
- 239000002356 single layer Substances 0.000 description 10
- 239000002904 solvent Substances 0.000 description 10
- IERHLVCPSMICTF-XVFCMESISA-N cytidine 5'-monophosphate Chemical group O=C1N=C(N)C=CN1[C@H]1[C@H](O)[C@H](O)[C@@H](COP(O)(O)=O)O1 IERHLVCPSMICTF-XVFCMESISA-N 0.000 description 9
- 239000000693 micelle Substances 0.000 description 9
- 230000002265 prevention Effects 0.000 description 9
- 239000000047 product Substances 0.000 description 9
- AUHDWARTFSKSAC-HEIFUQTGSA-N (2S,3R,4S,5R)-3,4-dihydroxy-5-(hydroxymethyl)-2-(6-oxo-1H-purin-9-yl)oxolane-2-carboxylic acid Chemical group [C@]1([C@H](O)[C@H](O)[C@@H](CO)O1)(N1C=NC=2C(O)=NC=NC12)C(=O)O AUHDWARTFSKSAC-HEIFUQTGSA-N 0.000 description 8
- HEDRZPFGACZZDS-UHFFFAOYSA-N Chloroform Chemical compound ClC(Cl)Cl HEDRZPFGACZZDS-UHFFFAOYSA-N 0.000 description 8
- 108091008874 T cell receptors Proteins 0.000 description 8
- 102000016266 T-Cell Antigen Receptors Human genes 0.000 description 8
- 102000002689 Toll-like receptor Human genes 0.000 description 8
- 108020000411 Toll-like receptor Proteins 0.000 description 8
- 230000027455 binding Effects 0.000 description 8
- OPTASPLRGRRNAP-UHFFFAOYSA-N cytosine Chemical compound NC=1C=CNC(=O)N=1 OPTASPLRGRRNAP-UHFFFAOYSA-N 0.000 description 8
- 208000035475 disorder Diseases 0.000 description 8
- 239000000839 emulsion Substances 0.000 description 8
- 210000000987 immune system Anatomy 0.000 description 8
- 230000035772 mutation Effects 0.000 description 8
- 239000002773 nucleotide Substances 0.000 description 8
- 125000003729 nucleotide group Chemical group 0.000 description 8
- 108091008875 B cell receptors Proteins 0.000 description 7
- 108090001030 Lipoproteins Proteins 0.000 description 7
- 102000004895 Lipoproteins Human genes 0.000 description 7
- KDXKERNSBIXSRK-UHFFFAOYSA-N Lysine Natural products NCCCCC(N)C(O)=O KDXKERNSBIXSRK-UHFFFAOYSA-N 0.000 description 7
- 230000000139 costimulatory effect Effects 0.000 description 7
- 150000004665 fatty acids Chemical class 0.000 description 7
- 239000000463 material Substances 0.000 description 7
- 230000004048 modification Effects 0.000 description 7
- 238000012986 modification Methods 0.000 description 7
- 102000004127 Cytokines Human genes 0.000 description 6
- 108090000695 Cytokines Proteins 0.000 description 6
- 102000014150 Interferons Human genes 0.000 description 6
- 108010050904 Interferons Proteins 0.000 description 6
- OKKJLVBELUTLKV-UHFFFAOYSA-N Methanol Chemical compound OC OKKJLVBELUTLKV-UHFFFAOYSA-N 0.000 description 6
- 108060008682 Tumor Necrosis Factor Proteins 0.000 description 6
- 102000000852 Tumor Necrosis Factor-alpha Human genes 0.000 description 6
- 229940127089 cytotoxic agent Drugs 0.000 description 6
- 230000000694 effects Effects 0.000 description 6
- 230000014509 gene expression Effects 0.000 description 6
- 230000001404 mediated effect Effects 0.000 description 6
- 230000008569 process Effects 0.000 description 6
- 229940002612 prodrug Drugs 0.000 description 6
- 239000000651 prodrug Substances 0.000 description 6
- 102000005962 receptors Human genes 0.000 description 6
- 108020003175 receptors Proteins 0.000 description 6
- 230000001954 sterilising effect Effects 0.000 description 6
- 238000004659 sterilization and disinfection Methods 0.000 description 6
- DMWMUMWKGKGSNW-OPMCLZTFSA-N (2S)-6-amino-2-[[(2S)-2-[[(2S)-6-amino-2-[[(2S)-2-[[(2S)-2-[[(2S,3S)-2-[[(2S)-4-amino-2-[[(2S)-2-[[(2S)-2-[[(2S)-2-[[(2S)-4-amino-2-[[(2S)-4-amino-2-[[2-[[(2R)-2-amino-3-[(2R)-2,3-di(hexadecanoyloxy)propyl]sulfanylpropanoyl]amino]acetyl]amino]-4-oxobutanoyl]amino]-4-oxobutanoyl]amino]-3-carboxypropanoyl]amino]-4-carboxybutanoyl]amino]-3-hydroxypropanoyl]amino]-4-oxobutanoyl]amino]-3-methylpentanoyl]amino]-3-hydroxypropanoyl]amino]-3-phenylpropanoyl]amino]hexanoyl]amino]-4-carboxybutanoyl]amino]hexanoic acid Chemical compound CCCCCCCCCCCCCCCC(=O)OC[C@H](CSC[C@H](N)C(=O)NCC(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CO)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@@H](CO)C(=O)N[C@@H](Cc1ccccc1)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CCCCN)C(O)=O)OC(=O)CCCCCCCCCCCCCCC DMWMUMWKGKGSNW-OPMCLZTFSA-N 0.000 description 5
- 241000894006 Bacteria Species 0.000 description 5
- 101710117490 Circumsporozoite protein Proteins 0.000 description 5
- 108010047041 Complementarity Determining Regions Proteins 0.000 description 5
- 241000588724 Escherichia coli Species 0.000 description 5
- GRSZFWQUAKGDAV-UHFFFAOYSA-N Inosinic acid Natural products OC1C(O)C(COP(O)(O)=O)OC1N1C(NC=NC2=O)=C2N=C1 GRSZFWQUAKGDAV-UHFFFAOYSA-N 0.000 description 5
- 102000015696 Interleukins Human genes 0.000 description 5
- 108010063738 Interleukins Proteins 0.000 description 5
- FBOZXECLQNJBKD-ZDUSSCGKSA-N L-methotrexate Chemical compound C=1N=C2N=C(N)N=C(N)C2=NC=1CN(C)C1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 FBOZXECLQNJBKD-ZDUSSCGKSA-N 0.000 description 5
- 108091054437 MHC class I family Proteins 0.000 description 5
- 241001465754 Metazoa Species 0.000 description 5
- 102100024216 Programmed cell death 1 ligand 1 Human genes 0.000 description 5
- 229930182558 Sterol Chemical class 0.000 description 5
- 230000003044 adaptive effect Effects 0.000 description 5
- 239000000556 agonist Substances 0.000 description 5
- 230000000890 antigenic effect Effects 0.000 description 5
- LMEKQMALGUDUQG-UHFFFAOYSA-N azathioprine Chemical compound CN1C=NC([N+]([O-])=O)=C1SC1=NC=NC2=C1NC=N2 LMEKQMALGUDUQG-UHFFFAOYSA-N 0.000 description 5
- 229960002170 azathioprine Drugs 0.000 description 5
- 239000002254 cytotoxic agent Substances 0.000 description 5
- 235000014113 dietary fatty acids Nutrition 0.000 description 5
- 239000003085 diluting agent Substances 0.000 description 5
- 229940023064 escherichia coli Drugs 0.000 description 5
- 229930195729 fatty acid Natural products 0.000 description 5
- 239000000194 fatty acid Substances 0.000 description 5
- 125000001924 fatty-acyl group Chemical group 0.000 description 5
- 150000004676 glycans Chemical class 0.000 description 5
- 229960001438 immunostimulant agent Drugs 0.000 description 5
- 230000001506 immunosuppresive effect Effects 0.000 description 5
- 230000021633 leukocyte mediated immunity Effects 0.000 description 5
- 239000007788 liquid Substances 0.000 description 5
- 201000004792 malaria Diseases 0.000 description 5
- 238000004519 manufacturing process Methods 0.000 description 5
- 229960000485 methotrexate Drugs 0.000 description 5
- 230000002441 reversible effect Effects 0.000 description 5
- 150000003432 sterols Chemical class 0.000 description 5
- 235000003702 sterols Nutrition 0.000 description 5
- YBJHBAHKTGYVGT-ZKWXMUAHSA-N (+)-Biotin Chemical compound N1C(=O)N[C@@H]2[C@H](CCCCC(=O)O)SC[C@@H]21 YBJHBAHKTGYVGT-ZKWXMUAHSA-N 0.000 description 4
- 108010071942 Colony-Stimulating Factors Proteins 0.000 description 4
- CMSMOCZEIVJLDB-UHFFFAOYSA-N Cyclophosphamide Chemical compound ClCCN(CCCl)P1(=O)NCCCO1 CMSMOCZEIVJLDB-UHFFFAOYSA-N 0.000 description 4
- 241000709661 Enterovirus Species 0.000 description 4
- 241000711549 Hepacivirus C Species 0.000 description 4
- 102100034458 Hepatitis A virus cellular receptor 2 Human genes 0.000 description 4
- 241000701044 Human gammaherpesvirus 4 Species 0.000 description 4
- UGQMRVRMYYASKQ-KQYNXXCUSA-N Inosine Chemical compound O[C@@H]1[C@H](O)[C@@H](CO)O[C@H]1N1C2=NC=NC(O)=C2N=C1 UGQMRVRMYYASKQ-KQYNXXCUSA-N 0.000 description 4
- 102000043129 MHC class I family Human genes 0.000 description 4
- 108700011259 MicroRNAs Proteins 0.000 description 4
- NWIBSHFKIJFRCO-WUDYKRTCSA-N Mytomycin Chemical compound C1N2C(C(C(C)=C(N)C3=O)=O)=C3[C@@H](COC(N)=O)[C@@]2(OC)[C@@H]2[C@H]1N2 NWIBSHFKIJFRCO-WUDYKRTCSA-N 0.000 description 4
- 241000283984 Rodentia Species 0.000 description 4
- 108091027967 Small hairpin RNA Proteins 0.000 description 4
- 230000024932 T cell mediated immunity Effects 0.000 description 4
- 230000003213 activating effect Effects 0.000 description 4
- 230000033289 adaptive immune response Effects 0.000 description 4
- 239000002246 antineoplastic agent Substances 0.000 description 4
- 239000008346 aqueous phase Substances 0.000 description 4
- 229960004397 cyclophosphamide Drugs 0.000 description 4
- 229940104302 cytosine Drugs 0.000 description 4
- 238000011161 development Methods 0.000 description 4
- 230000018109 developmental process Effects 0.000 description 4
- 230000006870 function Effects 0.000 description 4
- 239000003018 immunosuppressive agent Substances 0.000 description 4
- 238000001727 in vivo Methods 0.000 description 4
- 229940079322 interferon Drugs 0.000 description 4
- 239000002679 microRNA Substances 0.000 description 4
- 229920001282 polysaccharide Polymers 0.000 description 4
- 239000005017 polysaccharide Substances 0.000 description 4
- 239000000843 powder Substances 0.000 description 4
- ZAHRKKWIAAJSAO-UHFFFAOYSA-N rapamycin Natural products COCC(O)C(=C/C(C)C(=O)CC(OC(=O)C1CCCCN1C(=O)C(=O)C2(O)OC(CC(OC)C(=CC=CC=CC(C)CC(C)C(=O)C)C)CCC2C)C(C)CC3CCC(O)C(C3)OC)C ZAHRKKWIAAJSAO-UHFFFAOYSA-N 0.000 description 4
- 230000009467 reduction Effects 0.000 description 4
- 229920006395 saturated elastomer Polymers 0.000 description 4
- 229960002930 sirolimus Drugs 0.000 description 4
- 239000004055 small Interfering RNA Substances 0.000 description 4
- 238000002791 soaking Methods 0.000 description 4
- 241000894007 species Species 0.000 description 4
- 230000009870 specific binding Effects 0.000 description 4
- 238000003260 vortexing Methods 0.000 description 4
- WWUZIQQURGPMPG-UHFFFAOYSA-N (-)-D-erythro-Sphingosine Natural products CCCCCCCCCCCCCC=CC(O)C(N)CO WWUZIQQURGPMPG-UHFFFAOYSA-N 0.000 description 3
- 102000007644 Colony-Stimulating Factors Human genes 0.000 description 3
- PMATZTZNYRCHOR-CGLBZJNRSA-N Cyclosporin A Chemical compound CC[C@@H]1NC(=O)[C@H]([C@H](O)[C@H](C)C\C=C\C)N(C)C(=O)[C@H](C(C)C)N(C)C(=O)[C@H](CC(C)C)N(C)C(=O)[C@H](CC(C)C)N(C)C(=O)[C@@H](C)NC(=O)[C@H](C)NC(=O)[C@H](CC(C)C)N(C)C(=O)[C@H](C(C)C)NC(=O)[C@H](CC(C)C)N(C)C(=O)CN(C)C1=O PMATZTZNYRCHOR-CGLBZJNRSA-N 0.000 description 3
- 108010036949 Cyclosporine Proteins 0.000 description 3
- GHASVSINZRGABV-UHFFFAOYSA-N Fluorouracil Chemical compound FC1=CNC(=O)NC1=O GHASVSINZRGABV-UHFFFAOYSA-N 0.000 description 3
- 102000003886 Glycoproteins Human genes 0.000 description 3
- 108090000288 Glycoproteins Proteins 0.000 description 3
- 102100039620 Granulocyte-macrophage colony-stimulating factor Human genes 0.000 description 3
- 102000018713 Histocompatibility Antigens Class II Human genes 0.000 description 3
- 101001068133 Homo sapiens Hepatitis A virus cellular receptor 2 Proteins 0.000 description 3
- 229930010555 Inosine Natural products 0.000 description 3
- 108091054438 MHC class II family Proteins 0.000 description 3
- 241000699670 Mus sp. Species 0.000 description 3
- 108091036414 Polyinosinic:polycytidylic acid Proteins 0.000 description 3
- 102100024213 Programmed cell death 1 ligand 2 Human genes 0.000 description 3
- 101710194807 Protective antigen Proteins 0.000 description 3
- 238000012228 RNA interference-mediated gene silencing Methods 0.000 description 3
- 108010003723 Single-Domain Antibodies Proteins 0.000 description 3
- QJJXYPPXXYFBGM-LFZNUXCKSA-N Tacrolimus Chemical compound C1C[C@@H](O)[C@H](OC)C[C@@H]1\C=C(/C)[C@@H]1[C@H](C)[C@@H](O)CC(=O)[C@H](CC=C)/C=C(C)/C[C@H](C)C[C@H](OC)[C@H]([C@H](C[C@H]2C)OC)O[C@@]2(O)C(=O)C(=O)N2CCCC[C@H]2C(=O)O1 QJJXYPPXXYFBGM-LFZNUXCKSA-N 0.000 description 3
- 208000036142 Viral infection Diseases 0.000 description 3
- 239000002253 acid Substances 0.000 description 3
- 239000013543 active substance Substances 0.000 description 3
- 125000000129 anionic group Chemical class 0.000 description 3
- 229940121363 anti-inflammatory agent Drugs 0.000 description 3
- 239000002260 anti-inflammatory agent Substances 0.000 description 3
- 230000031018 biological processes and functions Effects 0.000 description 3
- 125000002091 cationic group Chemical class 0.000 description 3
- 229960004630 chlorambucil Drugs 0.000 description 3
- JCKYGMPEJWAADB-UHFFFAOYSA-N chlorambucil Chemical compound OC(=O)CCCC1=CC=C(N(CCCl)CCCl)C=C1 JCKYGMPEJWAADB-UHFFFAOYSA-N 0.000 description 3
- 229960001265 ciclosporin Drugs 0.000 description 3
- 229940047120 colony stimulating factors Drugs 0.000 description 3
- 229930182912 cyclosporin Natural products 0.000 description 3
- 231100000433 cytotoxic Toxicity 0.000 description 3
- 210000001151 cytotoxic T lymphocyte Anatomy 0.000 description 3
- 231100000599 cytotoxic agent Toxicity 0.000 description 3
- 230000001472 cytotoxic effect Effects 0.000 description 3
- 230000002708 enhancing effect Effects 0.000 description 3
- 230000008020 evaporation Effects 0.000 description 3
- 238000001704 evaporation Methods 0.000 description 3
- 238000001914 filtration Methods 0.000 description 3
- 229960002949 fluorouracil Drugs 0.000 description 3
- 230000009368 gene silencing by RNA Effects 0.000 description 3
- 239000011521 glass Substances 0.000 description 3
- 239000003102 growth factor Substances 0.000 description 3
- 230000028996 humoral immune response Effects 0.000 description 3
- 230000036571 hydration Effects 0.000 description 3
- 238000006703 hydration reaction Methods 0.000 description 3
- 229940072221 immunoglobulins Drugs 0.000 description 3
- 229940125721 immunosuppressive agent Drugs 0.000 description 3
- 229960003786 inosine Drugs 0.000 description 3
- 230000003993 interaction Effects 0.000 description 3
- 229940047122 interleukins Drugs 0.000 description 3
- 238000004255 ion exchange chromatography Methods 0.000 description 3
- 208000032839 leukemia Diseases 0.000 description 3
- 229960004961 mechlorethamine Drugs 0.000 description 3
- HAWPXGHAZFHHAD-UHFFFAOYSA-N mechlorethamine Chemical compound ClCCN(C)CCCl HAWPXGHAZFHHAD-UHFFFAOYSA-N 0.000 description 3
- 238000004007 reversed phase HPLC Methods 0.000 description 3
- 150000003384 small molecules Chemical class 0.000 description 3
- WWUZIQQURGPMPG-KRWOKUGFSA-N sphingosine Chemical compound CCCCCCCCCCCCC\C=C\[C@@H](O)[C@@H](N)CO WWUZIQQURGPMPG-KRWOKUGFSA-N 0.000 description 3
- 230000004936 stimulating effect Effects 0.000 description 3
- 208000024891 symptom Diseases 0.000 description 3
- 229960001967 tacrolimus Drugs 0.000 description 3
- QJJXYPPXXYFBGM-SHYZHZOCSA-N tacrolimus Natural products CO[C@H]1C[C@H](CC[C@@H]1O)C=C(C)[C@H]2OC(=O)[C@H]3CCCCN3C(=O)C(=O)[C@@]4(O)O[C@@H]([C@H](C[C@H]4C)OC)[C@@H](C[C@H](C)CC(=C[C@@H](CC=C)C(=O)C[C@H](O)[C@H]2C)C)OC QJJXYPPXXYFBGM-SHYZHZOCSA-N 0.000 description 3
- 230000001225 therapeutic effect Effects 0.000 description 3
- 238000002255 vaccination Methods 0.000 description 3
- 230000009385 viral infection Effects 0.000 description 3
- TZCPCKNHXULUIY-RGULYWFUSA-N 1,2-distearoyl-sn-glycero-3-phosphoserine Chemical compound CCCCCCCCCCCCCCCCCC(=O)OC[C@H](COP(O)(=O)OC[C@H](N)C(O)=O)OC(=O)CCCCCCCCCCCCCCCCC TZCPCKNHXULUIY-RGULYWFUSA-N 0.000 description 2
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 2
- 102100029822 B- and T-lymphocyte attenuator Human genes 0.000 description 2
- 101710144268 B- and T-lymphocyte attenuator Proteins 0.000 description 2
- 108010074708 B7-H1 Antigen Proteins 0.000 description 2
- 241000588832 Bordetella pertussis Species 0.000 description 2
- 206010006187 Breast cancer Diseases 0.000 description 2
- 208000026310 Breast neoplasm Diseases 0.000 description 2
- 241000589562 Brucella Species 0.000 description 2
- 102100027207 CD27 antigen Human genes 0.000 description 2
- 102100038078 CD276 antigen Human genes 0.000 description 2
- 101150013553 CD40 gene Proteins 0.000 description 2
- 241001493160 California encephalitis virus Species 0.000 description 2
- 241000222120 Candida <Saccharomycetales> Species 0.000 description 2
- DLGOEMSEDOSKAD-UHFFFAOYSA-N Carmustine Chemical compound ClCCNC(=O)N(N=O)CCCl DLGOEMSEDOSKAD-UHFFFAOYSA-N 0.000 description 2
- 108090000994 Catalytic RNA Proteins 0.000 description 2
- 102000053642 Catalytic RNA Human genes 0.000 description 2
- 108010012236 Chemokines Proteins 0.000 description 2
- 102000019034 Chemokines Human genes 0.000 description 2
- 241001647372 Chlamydia pneumoniae Species 0.000 description 2
- 241001647378 Chlamydia psittaci Species 0.000 description 2
- 206010008631 Cholera Diseases 0.000 description 2
- 241000193155 Clostridium botulinum Species 0.000 description 2
- 241000223205 Coccidioides immitis Species 0.000 description 2
- 241000711573 Coronaviridae Species 0.000 description 2
- 241000700626 Cowpox virus Species 0.000 description 2
- 241001337994 Cryptococcus <scale insect> Species 0.000 description 2
- XDTMQSROBMDMFD-UHFFFAOYSA-N Cyclohexane Chemical compound C1CCCCC1 XDTMQSROBMDMFD-UHFFFAOYSA-N 0.000 description 2
- 229930183912 Cytidylic acid Natural products 0.000 description 2
- 241000701022 Cytomegalovirus Species 0.000 description 2
- 241000725619 Dengue virus Species 0.000 description 2
- AOJJSUZBOXZQNB-TZSSRYMLSA-N Doxorubicin Chemical compound O([C@H]1C[C@@](O)(CC=2C(O)=C3C(=O)C=4C=CC=C(C=4C(=O)C3=C(O)C=21)OC)C(=O)CO)[C@H]1C[C@H](N)[C@H](O)[C@H](C)O1 AOJJSUZBOXZQNB-TZSSRYMLSA-N 0.000 description 2
- 241001115402 Ebolavirus Species 0.000 description 2
- 241000991587 Enterovirus C Species 0.000 description 2
- 102000003951 Erythropoietin Human genes 0.000 description 2
- 108090000394 Erythropoietin Proteins 0.000 description 2
- 241001646719 Escherichia coli O157:H7 Species 0.000 description 2
- HKVAMNSJSFKALM-GKUWKFKPSA-N Everolimus Chemical compound C1C[C@@H](OCCO)[C@H](OC)C[C@@H]1C[C@@H](C)[C@H]1OC(=O)[C@@H]2CCCCN2C(=O)C(=O)[C@](O)(O2)[C@H](C)CC[C@H]2C[C@H](OC)/C(C)=C/C=C/C=C/[C@@H](C)C[C@@H](C)C(=O)[C@H](OC)[C@H](O)/C(C)=C/[C@@H](C)C(=O)C1 HKVAMNSJSFKALM-GKUWKFKPSA-N 0.000 description 2
- 201000001342 Fallopian tube cancer Diseases 0.000 description 2
- 208000013452 Fallopian tube neoplasm Diseases 0.000 description 2
- 241000710831 Flavivirus Species 0.000 description 2
- 102000012673 Follicle Stimulating Hormone Human genes 0.000 description 2
- 108010079345 Follicle Stimulating Hormone Proteins 0.000 description 2
- ZWZWYGMENQVNFU-UHFFFAOYSA-N Glycerophosphorylserin Natural products OC(=O)C(N)COP(O)(=O)OCC(O)CO ZWZWYGMENQVNFU-UHFFFAOYSA-N 0.000 description 2
- 108010017080 Granulocyte Colony-Stimulating Factor Proteins 0.000 description 2
- 102000004269 Granulocyte Colony-Stimulating Factor Human genes 0.000 description 2
- 108010017213 Granulocyte-Macrophage Colony-Stimulating Factor Proteins 0.000 description 2
- 241000606768 Haemophilus influenzae Species 0.000 description 2
- 241000150562 Hantaan orthohantavirus Species 0.000 description 2
- 241000590002 Helicobacter pylori Species 0.000 description 2
- 241000700721 Hepatitis B virus Species 0.000 description 2
- 241000709721 Hepatovirus A Species 0.000 description 2
- 101000914511 Homo sapiens CD27 antigen Proteins 0.000 description 2
- 101000884279 Homo sapiens CD276 antigen Proteins 0.000 description 2
- 101001117317 Homo sapiens Programmed cell death 1 ligand 1 Proteins 0.000 description 2
- 101000914514 Homo sapiens T-cell-specific surface glycoprotein CD28 Proteins 0.000 description 2
- 101000801234 Homo sapiens Tumor necrosis factor receptor superfamily member 18 Proteins 0.000 description 2
- 101000955999 Homo sapiens V-set domain-containing T-cell activation inhibitor 1 Proteins 0.000 description 2
- 108010000521 Human Growth Hormone Proteins 0.000 description 2
- 102000002265 Human Growth Hormone Human genes 0.000 description 2
- 239000000854 Human Growth Hormone Substances 0.000 description 2
- 241000598171 Human adenovirus sp. Species 0.000 description 2
- 241000700588 Human alphaherpesvirus 1 Species 0.000 description 2
- 241000701074 Human alphaherpesvirus 2 Species 0.000 description 2
- 241001502974 Human gammaherpesvirus 8 Species 0.000 description 2
- 101800000324 Immunoglobulin A1 protease translocator Proteins 0.000 description 2
- 206010062016 Immunosuppression Diseases 0.000 description 2
- 241000712431 Influenza A virus Species 0.000 description 2
- 241000713196 Influenza B virus Species 0.000 description 2
- 241000713297 Influenza C virus Species 0.000 description 2
- 108090000174 Interleukin-10 Proteins 0.000 description 2
- UETNIIAIRMUTSM-UHFFFAOYSA-N Jacareubin Natural products CC1(C)OC2=CC3Oc4c(O)c(O)ccc4C(=O)C3C(=C2C=C1)O UETNIIAIRMUTSM-UHFFFAOYSA-N 0.000 description 2
- 241000710842 Japanese encephalitis virus Species 0.000 description 2
- 108010043610 KIR Receptors Proteins 0.000 description 2
- 102000002698 KIR Receptors Human genes 0.000 description 2
- 102000017578 LAG3 Human genes 0.000 description 2
- 241000589248 Legionella Species 0.000 description 2
- 208000007764 Legionnaires' Disease Diseases 0.000 description 2
- 241000589902 Leptospira Species 0.000 description 2
- 241000186781 Listeria Species 0.000 description 2
- 102000009151 Luteinizing Hormone Human genes 0.000 description 2
- 108010073521 Luteinizing Hormone Proteins 0.000 description 2
- 241000712899 Lymphocytic choriomeningitis mammarenavirus Species 0.000 description 2
- 239000004472 Lysine Substances 0.000 description 2
- 241000124008 Mammalia Species 0.000 description 2
- 241000712079 Measles morbillivirus Species 0.000 description 2
- 241000579048 Merkel cell polyomavirus Species 0.000 description 2
- 241000711386 Mumps virus Species 0.000 description 2
- 101100407308 Mus musculus Pdcd1lg2 gene Proteins 0.000 description 2
- 241000186359 Mycobacterium Species 0.000 description 2
- 241000202934 Mycoplasma pneumoniae Species 0.000 description 2
- PXHVJJICTQNCMI-UHFFFAOYSA-N Nickel Chemical compound [Ni] PXHVJJICTQNCMI-UHFFFAOYSA-N 0.000 description 2
- 241001263478 Norovirus Species 0.000 description 2
- 101710163270 Nuclease Proteins 0.000 description 2
- 108010038807 Oligopeptides Proteins 0.000 description 2
- 102000015636 Oligopeptides Human genes 0.000 description 2
- 241000702244 Orthoreovirus Species 0.000 description 2
- 206010033128 Ovarian cancer Diseases 0.000 description 2
- 206010061535 Ovarian neoplasm Diseases 0.000 description 2
- 229910019142 PO4 Inorganic materials 0.000 description 2
- 208000002606 Paramyxoviridae Infections Diseases 0.000 description 2
- 201000005702 Pertussis Diseases 0.000 description 2
- NBIIXXVUZAFLBC-UHFFFAOYSA-N Phosphoric acid Chemical compound OP(O)(O)=O NBIIXXVUZAFLBC-UHFFFAOYSA-N 0.000 description 2
- 206010035226 Plasma cell myeloma Diseases 0.000 description 2
- 241000223960 Plasmodium falciparum Species 0.000 description 2
- 241000223801 Plasmodium knowlesi Species 0.000 description 2
- 241000223821 Plasmodium malariae Species 0.000 description 2
- 241001505293 Plasmodium ovale Species 0.000 description 2
- 241000223810 Plasmodium vivax Species 0.000 description 2
- 206010035664 Pneumonia Diseases 0.000 description 2
- 241000711902 Pneumovirus Species 0.000 description 2
- 241001505332 Polyomavirus sp. Species 0.000 description 2
- 108700030875 Programmed Cell Death 1 Ligand 2 Proteins 0.000 description 2
- 102100040678 Programmed cell death protein 1 Human genes 0.000 description 2
- 206010060862 Prostate cancer Diseases 0.000 description 2
- 208000000236 Prostatic Neoplasms Diseases 0.000 description 2
- 241000125945 Protoparvovirus Species 0.000 description 2
- 241000621172 Pseudocowpox virus Species 0.000 description 2
- 241000711798 Rabies lyssavirus Species 0.000 description 2
- 108010008281 Recombinant Fusion Proteins Proteins 0.000 description 2
- 102000007056 Recombinant Fusion Proteins Human genes 0.000 description 2
- 241000702670 Rotavirus Species 0.000 description 2
- 241000710799 Rubella virus Species 0.000 description 2
- 241000607142 Salmonella Species 0.000 description 2
- 241000837158 Senecavirus A Species 0.000 description 2
- MTCFGRXMJLQNBG-UHFFFAOYSA-N Serine Natural products OCC(N)C(O)=O MTCFGRXMJLQNBG-UHFFFAOYSA-N 0.000 description 2
- 241000607768 Shigella Species 0.000 description 2
- 101710200413 Small hydrophobic protein Proteins 0.000 description 2
- 241000191940 Staphylococcus Species 0.000 description 2
- 241000193998 Streptococcus pneumoniae Species 0.000 description 2
- 102100027213 T-cell-specific surface glycoprotein CD28 Human genes 0.000 description 2
- 108010055044 Tetanus Toxin Proteins 0.000 description 2
- 102000036693 Thrombopoietin Human genes 0.000 description 2
- 108010041111 Thrombopoietin Proteins 0.000 description 2
- 102000011923 Thyrotropin Human genes 0.000 description 2
- 108010061174 Thyrotropin Proteins 0.000 description 2
- 108010009583 Transforming Growth Factors Proteins 0.000 description 2
- 102000009618 Transforming Growth Factors Human genes 0.000 description 2
- 102100040245 Tumor necrosis factor receptor superfamily member 5 Human genes 0.000 description 2
- DRTQHJPVMGBUCF-XVFCMESISA-N Uridine Chemical compound O[C@@H]1[C@H](O)[C@@H](CO)O[C@H]1N1C(=O)NC(=O)C=C1 DRTQHJPVMGBUCF-XVFCMESISA-N 0.000 description 2
- 102100038929 V-set domain-containing T-cell activation inhibitor 1 Human genes 0.000 description 2
- 241000700618 Vaccinia virus Species 0.000 description 2
- 241000710886 West Nile virus Species 0.000 description 2
- 241000710772 Yellow fever virus Species 0.000 description 2
- 241000607447 Yersinia enterocolitica Species 0.000 description 2
- LUXUAZKGQZPOBZ-SAXJAHGMSA-N [(3S,4S,5S,6R)-3,4,5-trihydroxy-6-(hydroxymethyl)oxan-2-yl] (Z)-octadec-9-enoate Chemical group CCCCCCCC\C=C/CCCCCCCC(=O)OC1O[C@H](CO)[C@@H](O)[C@H](O)[C@@H]1O LUXUAZKGQZPOBZ-SAXJAHGMSA-N 0.000 description 2
- RJURFGZVJUQBHK-UHFFFAOYSA-N actinomycin D Natural products CC1OC(=O)C(C(C)C)N(C)C(=O)CN(C)C(=O)C2CCCN2C(=O)C(C(C)C)NC(=O)C1NC(=O)C1=C(N)C(=O)C(C)=C2OC(C(C)=CC=C3C(=O)NC4C(=O)NC(C(N5CCCC5C(=O)N(C)CC(=O)N(C)C(C(C)C)C(=O)OC4C)=O)C(C)C)=C3N=C21 RJURFGZVJUQBHK-UHFFFAOYSA-N 0.000 description 2
- 229960002964 adalimumab Drugs 0.000 description 2
- 125000000539 amino acid group Chemical group 0.000 description 2
- 230000000692 anti-sense effect Effects 0.000 description 2
- 230000009286 beneficial effect Effects 0.000 description 2
- 230000008901 benefit Effects 0.000 description 2
- 230000015572 biosynthetic process Effects 0.000 description 2
- 229960002685 biotin Drugs 0.000 description 2
- 235000020958 biotin Nutrition 0.000 description 2
- 239000011616 biotin Substances 0.000 description 2
- 239000003153 chemical reaction reagent Substances 0.000 description 2
- 235000018417 cysteine Nutrition 0.000 description 2
- XUJNEKJLAYXESH-UHFFFAOYSA-N cysteine Natural products SCC(N)C(O)=O XUJNEKJLAYXESH-UHFFFAOYSA-N 0.000 description 2
- 125000000151 cysteine group Chemical group N[C@@H](CS)C(=O)* 0.000 description 2
- IERHLVCPSMICTF-UHFFFAOYSA-N cytidine monophosphate Natural products O=C1N=C(N)C=CN1C1C(O)C(O)C(COP(O)(O)=O)O1 IERHLVCPSMICTF-UHFFFAOYSA-N 0.000 description 2
- 210000004443 dendritic cell Anatomy 0.000 description 2
- 229960003957 dexamethasone Drugs 0.000 description 2
- UREBDLICKHMUKA-CXSFZGCWSA-N dexamethasone Chemical compound C1CC2=CC(=O)C=C[C@]2(C)[C@]2(F)[C@@H]1[C@@H]1C[C@@H](C)[C@@](C(=O)CO)(O)[C@@]1(C)C[C@@H]2O UREBDLICKHMUKA-CXSFZGCWSA-N 0.000 description 2
- 150000001982 diacylglycerols Chemical class 0.000 description 2
- 206010013023 diphtheria Diseases 0.000 description 2
- 239000006185 dispersion Substances 0.000 description 2
- 238000009826 distribution Methods 0.000 description 2
- 229940079593 drug Drugs 0.000 description 2
- 239000003995 emulsifying agent Substances 0.000 description 2
- 239000002158 endotoxin Substances 0.000 description 2
- 238000005516 engineering process Methods 0.000 description 2
- 230000000688 enterotoxigenic effect Effects 0.000 description 2
- 229940105423 erythropoietin Drugs 0.000 description 2
- 229960005167 everolimus Drugs 0.000 description 2
- 229960000556 fingolimod Drugs 0.000 description 2
- KKGQTZUTZRNORY-UHFFFAOYSA-N fingolimod Chemical compound CCCCCCCCC1=CC=C(CCC(N)(CO)CO)C=C1 KKGQTZUTZRNORY-UHFFFAOYSA-N 0.000 description 2
- 239000012530 fluid Substances 0.000 description 2
- 229940028334 follicle stimulating hormone Drugs 0.000 description 2
- 238000009472 formulation Methods 0.000 description 2
- 230000008014 freezing Effects 0.000 description 2
- 238000007710 freezing Methods 0.000 description 2
- 208000005017 glioblastoma Diseases 0.000 description 2
- 229940047650 haemophilus influenzae Drugs 0.000 description 2
- 230000003394 haemopoietic effect Effects 0.000 description 2
- 229940037467 helicobacter pylori Drugs 0.000 description 2
- 210000002443 helper t lymphocyte Anatomy 0.000 description 2
- 239000008240 homogeneous mixture Substances 0.000 description 2
- 229940088597 hormone Drugs 0.000 description 2
- 239000005556 hormone Substances 0.000 description 2
- 150000002430 hydrocarbons Chemical group 0.000 description 2
- 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 description 2
- 210000002865 immune cell Anatomy 0.000 description 2
- 230000036039 immunity Effects 0.000 description 2
- 230000002163 immunogen Effects 0.000 description 2
- 230000003308 immunostimulating effect Effects 0.000 description 2
- 230000001976 improved effect Effects 0.000 description 2
- 239000012535 impurity Substances 0.000 description 2
- 238000010348 incorporation Methods 0.000 description 2
- CGIGDMFJXJATDK-UHFFFAOYSA-N indomethacin Chemical compound CC1=C(CC(O)=O)C2=CC(OC)=CC=C2N1C(=O)C1=CC=C(Cl)C=C1 CGIGDMFJXJATDK-UHFFFAOYSA-N 0.000 description 2
- 239000012678 infectious agent Substances 0.000 description 2
- 230000002757 inflammatory effect Effects 0.000 description 2
- 229960000598 infliximab Drugs 0.000 description 2
- 239000003112 inhibitor Substances 0.000 description 2
- 150000002484 inorganic compounds Chemical class 0.000 description 2
- 229910010272 inorganic material Inorganic materials 0.000 description 2
- 229940028843 inosinic acid Drugs 0.000 description 2
- 235000013902 inosinic acid Nutrition 0.000 description 2
- 239000004245 inosinic acid Substances 0.000 description 2
- NOESYZHRGYRDHS-UHFFFAOYSA-N insulin Chemical compound N1C(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(NC(=O)CN)C(C)CC)CSSCC(C(NC(CO)C(=O)NC(CC(C)C)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CCC(N)=O)C(=O)NC(CC(C)C)C(=O)NC(CCC(O)=O)C(=O)NC(CC(N)=O)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CSSCC(NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2C=CC(O)=CC=2)NC(=O)C(CC(C)C)NC(=O)C(C)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2NC=NC=2)NC(=O)C(CO)NC(=O)CNC2=O)C(=O)NCC(=O)NC(CCC(O)=O)C(=O)NC(CCCNC(N)=N)C(=O)NCC(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC(O)=CC=3)C(=O)NC(C(C)O)C(=O)N3C(CCC3)C(=O)NC(CCCCN)C(=O)NC(C)C(O)=O)C(=O)NC(CC(N)=O)C(O)=O)=O)NC(=O)C(C(C)CC)NC(=O)C(CO)NC(=O)C(C(C)O)NC(=O)C1CSSCC2NC(=O)C(CC(C)C)NC(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CC(N)=O)NC(=O)C(NC(=O)C(N)CC=1C=CC=CC=1)C(C)C)CC1=CN=CN1 NOESYZHRGYRDHS-UHFFFAOYSA-N 0.000 description 2
- 229940047124 interferons Drugs 0.000 description 2
- 239000003446 ligand Substances 0.000 description 2
- 230000000670 limiting effect Effects 0.000 description 2
- 229940040129 luteinizing hormone Drugs 0.000 description 2
- 230000014759 maintenance of location Effects 0.000 description 2
- 108020004999 messenger RNA Proteins 0.000 description 2
- 206010061289 metastatic neoplasm Diseases 0.000 description 2
- 125000002496 methyl group Chemical group [H]C([H])([H])* 0.000 description 2
- 229960004857 mitomycin Drugs 0.000 description 2
- 125000001419 myristoyl group Chemical group O=C([*])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])[H] 0.000 description 2
- 229910052757 nitrogen Inorganic materials 0.000 description 2
- 239000000041 non-steroidal anti-inflammatory agent Substances 0.000 description 2
- 229940021182 non-steroidal anti-inflammatory drug Drugs 0.000 description 2
- 102000039446 nucleic acids Human genes 0.000 description 2
- 108020004707 nucleic acids Proteins 0.000 description 2
- 150000007523 nucleic acids Chemical class 0.000 description 2
- 239000007764 o/w emulsion Substances 0.000 description 2
- 150000002894 organic compounds Chemical class 0.000 description 2
- 125000001312 palmitoyl group Chemical group O=C([*])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])[H] 0.000 description 2
- 244000045947 parasite Species 0.000 description 2
- 201000002628 peritoneum cancer Diseases 0.000 description 2
- WTJKGGKOPKCXLL-RRHRGVEJSA-N phosphatidylcholine Chemical compound CCCCCCCCCCCCCCCC(=O)OC[C@H](COP([O-])(=O)OCC[N+](C)(C)C)OC(=O)CCCCCCCC=CCCCCCCCC WTJKGGKOPKCXLL-RRHRGVEJSA-N 0.000 description 2
- YHHSONZFOIEMCP-UHFFFAOYSA-O phosphocholine Chemical compound C[N+](C)(C)CCOP(O)(O)=O YHHSONZFOIEMCP-UHFFFAOYSA-O 0.000 description 2
- 230000004962 physiological condition Effects 0.000 description 2
- 229940118768 plasmodium malariae Drugs 0.000 description 2
- 229920000642 polymer Polymers 0.000 description 2
- OXCMYAYHXIHQOA-UHFFFAOYSA-N potassium;[2-butyl-5-chloro-3-[[4-[2-(1,2,4-triaza-3-azanidacyclopenta-1,4-dien-5-yl)phenyl]phenyl]methyl]imidazol-4-yl]methanol Chemical compound [K+].CCCCC1=NC(Cl)=C(CO)N1CC1=CC=C(C=2C(=CC=CC=2)C2=N[N-]N=N2)C=C1 OXCMYAYHXIHQOA-UHFFFAOYSA-N 0.000 description 2
- 230000003389 potentiating effect Effects 0.000 description 2
- XOFYZVNMUHMLCC-ZPOLXVRWSA-N prednisone Chemical compound O=C1C=C[C@]2(C)[C@H]3C(=O)C[C@](C)([C@@](CC4)(O)C(=O)CO)[C@@H]4[C@@H]3CCC2=C1 XOFYZVNMUHMLCC-ZPOLXVRWSA-N 0.000 description 2
- 229960004618 prednisone Drugs 0.000 description 2
- 230000002035 prolonged effect Effects 0.000 description 2
- 230000004044 response Effects 0.000 description 2
- PYWVYCXTNDRMGF-UHFFFAOYSA-N rhodamine B Chemical compound [Cl-].C=12C=CC(=[N+](CC)CC)C=C2OC2=CC(N(CC)CC)=CC=C2C=1C1=CC=CC=C1C(O)=O PYWVYCXTNDRMGF-UHFFFAOYSA-N 0.000 description 2
- 108091092562 ribozyme Proteins 0.000 description 2
- WVYADZUPLLSGPU-UHFFFAOYSA-N salsalate Chemical compound OC(=O)C1=CC=CC=C1OC(=O)C1=CC=CC=C1O WVYADZUPLLSGPU-UHFFFAOYSA-N 0.000 description 2
- 210000002966 serum Anatomy 0.000 description 2
- 230000000087 stabilizing effect Effects 0.000 description 2
- 125000003696 stearoyl group Chemical group O=C([*])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])[H] 0.000 description 2
- 239000011550 stock solution Substances 0.000 description 2
- 229940031000 streptococcus pneumoniae Drugs 0.000 description 2
- 229960001940 sulfasalazine Drugs 0.000 description 2
- NCEXYHBECQHGNR-QZQOTICOSA-N sulfasalazine Chemical compound C1=C(O)C(C(=O)O)=CC(\N=N\C=2C=CC(=CC=2)S(=O)(=O)NC=2N=CC=CC=2)=C1 NCEXYHBECQHGNR-QZQOTICOSA-N 0.000 description 2
- NCEXYHBECQHGNR-UHFFFAOYSA-N sulfasalazine Natural products C1=C(O)C(C(=O)O)=CC(N=NC=2C=CC(=CC=2)S(=O)(=O)NC=2N=CC=CC=2)=C1 NCEXYHBECQHGNR-UHFFFAOYSA-N 0.000 description 2
- 230000001629 suppression Effects 0.000 description 2
- 229940118376 tetanus toxin Drugs 0.000 description 2
- WYWHKKSPHMUBEB-UHFFFAOYSA-N tioguanine Chemical compound N1C(N)=NC(=S)C2=C1N=CN2 WYWHKKSPHMUBEB-UHFFFAOYSA-N 0.000 description 2
- 230000009261 transgenic effect Effects 0.000 description 2
- 241000712461 unidentified influenza virus Species 0.000 description 2
- 239000007762 w/o emulsion Substances 0.000 description 2
- 229940051021 yellow-fever virus Drugs 0.000 description 2
- 229940098232 yersinia enterocolitica Drugs 0.000 description 2
- RDJGLLICXDHJDY-NSHDSACASA-N (2s)-2-(3-phenoxyphenyl)propanoic acid Chemical compound OC(=O)[C@@H](C)C1=CC=CC(OC=2C=CC=CC=2)=C1 RDJGLLICXDHJDY-NSHDSACASA-N 0.000 description 1
- LJRDOKAZOAKLDU-UDXJMMFXSA-N (2s,3s,4r,5r,6r)-5-amino-2-(aminomethyl)-6-[(2r,3s,4r,5s)-5-[(1r,2r,3s,5r,6s)-3,5-diamino-2-[(2s,3r,4r,5s,6r)-3-amino-4,5-dihydroxy-6-(hydroxymethyl)oxan-2-yl]oxy-6-hydroxycyclohexyl]oxy-4-hydroxy-2-(hydroxymethyl)oxolan-3-yl]oxyoxane-3,4-diol;sulfuric ac Chemical compound OS(O)(=O)=O.N[C@@H]1[C@@H](O)[C@H](O)[C@H](CN)O[C@@H]1O[C@H]1[C@@H](O)[C@H](O[C@H]2[C@@H]([C@@H](N)C[C@@H](N)[C@@H]2O)O[C@@H]2[C@@H]([C@@H](O)[C@H](O)[C@@H](CO)O2)N)O[C@@H]1CO LJRDOKAZOAKLDU-UDXJMMFXSA-N 0.000 description 1
- YPBKTZBXSBLTDK-PKNBQFBNSA-N (3e)-3-[(3-bromo-4-fluoroanilino)-nitrosomethylidene]-4-[2-(sulfamoylamino)ethylamino]-1,2,5-oxadiazole Chemical compound NS(=O)(=O)NCCNC1=NON\C1=C(N=O)/NC1=CC=C(F)C(Br)=C1 YPBKTZBXSBLTDK-PKNBQFBNSA-N 0.000 description 1
- MZOFCQQQCNRIBI-VMXHOPILSA-N (3s)-4-[[(2s)-1-[[(2s)-1-[[(1s)-1-carboxy-2-hydroxyethyl]amino]-4-methyl-1-oxopentan-2-yl]amino]-5-(diaminomethylideneamino)-1-oxopentan-2-yl]amino]-3-[[2-[[(2s)-2,6-diaminohexanoyl]amino]acetyl]amino]-4-oxobutanoic acid Chemical compound OC[C@@H](C(O)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CCCN=C(N)N)NC(=O)[C@H](CC(O)=O)NC(=O)CNC(=O)[C@@H](N)CCCCN MZOFCQQQCNRIBI-VMXHOPILSA-N 0.000 description 1
- OMJKFYKNWZZKTK-POHAHGRESA-N (5z)-5-(dimethylaminohydrazinylidene)imidazole-4-carboxamide Chemical compound CN(C)N\N=C1/N=CN=C1C(N)=O OMJKFYKNWZZKTK-POHAHGRESA-N 0.000 description 1
- HMLGSIZOMSVISS-ONJSNURVSA-N (7r)-7-[[(2z)-2-(2-amino-1,3-thiazol-4-yl)-2-(2,2-dimethylpropanoyloxymethoxyimino)acetyl]amino]-3-ethenyl-8-oxo-5-thia-1-azabicyclo[4.2.0]oct-2-ene-2-carboxylic acid Chemical compound N([C@@H]1C(N2C(=C(C=C)CSC21)C(O)=O)=O)C(=O)\C(=N/OCOC(=O)C(C)(C)C)C1=CSC(N)=N1 HMLGSIZOMSVISS-ONJSNURVSA-N 0.000 description 1
- WHTVZRBIWZFKQO-AWEZNQCLSA-N (S)-chloroquine Chemical compound ClC1=CC=C2C(N[C@@H](C)CCCN(CC)CC)=CC=NC2=C1 WHTVZRBIWZFKQO-AWEZNQCLSA-N 0.000 description 1
- 102000040650 (ribonucleotides)n+m Human genes 0.000 description 1
- ZORQXIQZAOLNGE-UHFFFAOYSA-N 1,1-difluorocyclohexane Chemical compound FC1(F)CCCCC1 ZORQXIQZAOLNGE-UHFFFAOYSA-N 0.000 description 1
- UEJJHQNACJXSKW-UHFFFAOYSA-N 2-(2,6-dioxopiperidin-3-yl)-1H-isoindole-1,3(2H)-dione Chemical compound O=C1C2=CC=CC=C2C(=O)N1C1CCC(=O)NC1=O UEJJHQNACJXSKW-UHFFFAOYSA-N 0.000 description 1
- OTLLEIBWKHEHGU-UHFFFAOYSA-N 2-[5-[[5-(6-aminopurin-9-yl)-3,4-dihydroxyoxolan-2-yl]methoxy]-3,4-dihydroxy-6-(hydroxymethyl)oxan-2-yl]oxy-3,5-dihydroxy-4-phosphonooxyhexanedioic acid Chemical compound C1=NC=2C(N)=NC=NC=2N1C(C(C1O)O)OC1COC1C(CO)OC(OC(C(O)C(OP(O)(O)=O)C(O)C(O)=O)C(O)=O)C(O)C1O OTLLEIBWKHEHGU-UHFFFAOYSA-N 0.000 description 1
- FSPQCTGGIANIJZ-UHFFFAOYSA-N 2-[[(3,4-dimethoxyphenyl)-oxomethyl]amino]-4,5,6,7-tetrahydro-1-benzothiophene-3-carboxamide Chemical compound C1=C(OC)C(OC)=CC=C1C(=O)NC1=C(C(N)=O)C(CCCC2)=C2S1 FSPQCTGGIANIJZ-UHFFFAOYSA-N 0.000 description 1
- AOJJSUZBOXZQNB-VTZDEGQISA-N 4'-epidoxorubicin Chemical compound O([C@H]1C[C@@](O)(CC=2C(O)=C3C(=O)C=4C=CC=C(C=4C(=O)C3=C(O)C=21)OC)C(=O)CO)[C@H]1C[C@H](N)[C@@H](O)[C@H](C)O1 AOJJSUZBOXZQNB-VTZDEGQISA-N 0.000 description 1
- RANONBLIHMVXAJ-UHFFFAOYSA-N 4-hydroxycyclophosphamide Chemical compound OC1CCOP(=O)(N(CCCl)CCCl)N1 RANONBLIHMVXAJ-UHFFFAOYSA-N 0.000 description 1
- IDPUKCWIGUEADI-UHFFFAOYSA-N 5-[bis(2-chloroethyl)amino]uracil Chemical compound ClCCN(CCCl)C1=CNC(=O)NC1=O IDPUKCWIGUEADI-UHFFFAOYSA-N 0.000 description 1
- MJZJYWCQPMNPRM-UHFFFAOYSA-N 6,6-dimethyl-1-[3-(2,4,5-trichlorophenoxy)propoxy]-1,6-dihydro-1,3,5-triazine-2,4-diamine Chemical compound CC1(C)N=C(N)N=C(N)N1OCCCOC1=CC(Cl)=C(Cl)C=C1Cl MJZJYWCQPMNPRM-UHFFFAOYSA-N 0.000 description 1
- STQGQHZAVUOBTE-UHFFFAOYSA-N 7-Cyan-hept-2t-en-4,6-diinsaeure Natural products C1=2C(O)=C3C(=O)C=4C(OC)=CC=CC=4C(=O)C3=C(O)C=2CC(O)(C(C)=O)CC1OC1CC(N)C(O)C(C)O1 STQGQHZAVUOBTE-UHFFFAOYSA-N 0.000 description 1
- 206010069754 Acquired gene mutation Diseases 0.000 description 1
- 241000251468 Actinopterygii Species 0.000 description 1
- 108010059616 Activins Proteins 0.000 description 1
- 102000005606 Activins Human genes 0.000 description 1
- 101150051188 Adora2a gene Proteins 0.000 description 1
- 102400000068 Angiostatin Human genes 0.000 description 1
- 108010079709 Angiostatins Proteins 0.000 description 1
- 108010005853 Anti-Mullerian Hormone Proteins 0.000 description 1
- 108020005544 Antisense RNA Proteins 0.000 description 1
- BSYNRYMUTXBXSQ-UHFFFAOYSA-N Aspirin Chemical compound CC(=O)OC1=CC=CC=C1C(O)=O BSYNRYMUTXBXSQ-UHFFFAOYSA-N 0.000 description 1
- XHVAWZZCDCWGBK-WYRLRVFGSA-M Aurothioglucose Chemical compound OC[C@H]1O[C@H](S[Au])[C@H](O)[C@@H](O)[C@@H]1O XHVAWZZCDCWGBK-WYRLRVFGSA-M 0.000 description 1
- 108091007065 BIRCs Proteins 0.000 description 1
- 102000052609 BRCA2 Human genes 0.000 description 1
- 108700020462 BRCA2 Proteins 0.000 description 1
- 208000035143 Bacterial infection Diseases 0.000 description 1
- 241000537222 Betabaculovirus Species 0.000 description 1
- 241000190944 Blastochloris viridis Species 0.000 description 1
- 108010006654 Bleomycin Proteins 0.000 description 1
- 101150008921 Brca2 gene Proteins 0.000 description 1
- COVZYZSDYWQREU-UHFFFAOYSA-N Busulfan Chemical compound CS(=O)(=O)OCCCCOS(C)(=O)=O COVZYZSDYWQREU-UHFFFAOYSA-N 0.000 description 1
- 102100032528 C-type lectin domain family 11 member A Human genes 0.000 description 1
- 101710167766 C-type lectin domain family 11 member A Proteins 0.000 description 1
- YDNKGFDKKRUKPY-JHOUSYSJSA-N C16 ceramide Natural products CCCCCCCCCCCCCCCC(=O)N[C@@H](CO)[C@H](O)C=CCCCCCCCCCCCCC YDNKGFDKKRUKPY-JHOUSYSJSA-N 0.000 description 1
- 102100024263 CD160 antigen Human genes 0.000 description 1
- 102100038077 CD226 antigen Human genes 0.000 description 1
- 102100025221 CD70 antigen Human genes 0.000 description 1
- 210000001266 CD8-positive T-lymphocyte Anatomy 0.000 description 1
- 210000001239 CD8-positive, alpha-beta cytotoxic T lymphocyte Anatomy 0.000 description 1
- 108010021064 CTLA-4 Antigen Proteins 0.000 description 1
- 229940045513 CTLA4 antagonist Drugs 0.000 description 1
- 101100496968 Caenorhabditis elegans ctc-1 gene Proteins 0.000 description 1
- KLWPJMFMVPTNCC-UHFFFAOYSA-N Camptothecin Natural products CCC1(O)C(=O)OCC2=C1C=C3C4Nc5ccccc5C=C4CN3C2=O KLWPJMFMVPTNCC-UHFFFAOYSA-N 0.000 description 1
- 101710167800 Capsid assembly scaffolding protein Proteins 0.000 description 1
- 101710132601 Capsid protein Proteins 0.000 description 1
- 229920002134 Carboxymethyl cellulose Polymers 0.000 description 1
- 201000009030 Carcinoma Diseases 0.000 description 1
- 102000020313 Cell-Penetrating Peptides Human genes 0.000 description 1
- 108010051109 Cell-Penetrating Peptides Proteins 0.000 description 1
- 206010008342 Cervix carcinoma Diseases 0.000 description 1
- 102100021809 Chorionic somatomammotropin hormone 1 Human genes 0.000 description 1
- UHDGCWIWMRVCDJ-CCXZUQQUSA-N Cytarabine Chemical compound O=C1N=C(N)C=CN1[C@H]1[C@@H](O)[C@H](O)[C@@H](CO)O1 UHDGCWIWMRVCDJ-CCXZUQQUSA-N 0.000 description 1
- 102100039498 Cytotoxic T-lymphocyte protein 4 Human genes 0.000 description 1
- XUIIKFGFIJCVMT-GFCCVEGCSA-N D-thyroxine Chemical compound IC1=CC(C[C@@H](N)C(O)=O)=CC(I)=C1OC1=CC(I)=C(O)C(I)=C1 XUIIKFGFIJCVMT-GFCCVEGCSA-N 0.000 description 1
- 230000033616 DNA repair Effects 0.000 description 1
- 108010092160 Dactinomycin Proteins 0.000 description 1
- 108700022150 Designed Ankyrin Repeat Proteins Proteins 0.000 description 1
- 241000255925 Diptera Species 0.000 description 1
- 206010061818 Disease progression Diseases 0.000 description 1
- XXPXYPLPSDPERN-UHFFFAOYSA-N Ecteinascidin 743 Natural products COc1cc2C(NCCc2cc1O)C(=O)OCC3N4C(O)C5Cc6cc(C)c(OC)c(O)c6C(C4C(S)c7c(OC(=O)C)c(C)c8OCOc8c37)N5C XXPXYPLPSDPERN-UHFFFAOYSA-N 0.000 description 1
- 102400001047 Endostatin Human genes 0.000 description 1
- 108010079505 Endostatins Proteins 0.000 description 1
- HTIJFSOGRVMCQR-UHFFFAOYSA-N Epirubicin Natural products COc1cccc2C(=O)c3c(O)c4CC(O)(CC(OC5CC(N)C(=O)C(C)O5)c4c(O)c3C(=O)c12)C(=O)CO HTIJFSOGRVMCQR-UHFFFAOYSA-N 0.000 description 1
- 108010008165 Etanercept Proteins 0.000 description 1
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 1
- 241000282326 Felis catus Species 0.000 description 1
- 102000018233 Fibroblast Growth Factor Human genes 0.000 description 1
- 108050007372 Fibroblast Growth Factor Proteins 0.000 description 1
- 238000005033 Fourier transform infrared spectroscopy Methods 0.000 description 1
- 108091006027 G proteins Proteins 0.000 description 1
- 108091000058 GTP-Binding Proteins 0.000 description 1
- 102100031351 Galectin-9 Human genes 0.000 description 1
- 101100229077 Gallus gallus GAL9 gene Proteins 0.000 description 1
- 244000068988 Glycine max Species 0.000 description 1
- 235000010469 Glycine max Nutrition 0.000 description 1
- 102000006771 Gonadotropins Human genes 0.000 description 1
- 108010086677 Gonadotropins Proteins 0.000 description 1
- 108060003393 Granulin Proteins 0.000 description 1
- 108010007707 Hepatitis A Virus Cellular Receptor 2 Proteins 0.000 description 1
- 102000008949 Histocompatibility Antigens Class I Human genes 0.000 description 1
- 241000282412 Homo Species 0.000 description 1
- 101000761938 Homo sapiens CD160 antigen Proteins 0.000 description 1
- 101000884298 Homo sapiens CD226 antigen Proteins 0.000 description 1
- 101000934356 Homo sapiens CD70 antigen Proteins 0.000 description 1
- 101001037256 Homo sapiens Indoleamine 2,3-dioxygenase 1 Proteins 0.000 description 1
- 101001037261 Homo sapiens Indoleamine 2,3-dioxygenase 2 Proteins 0.000 description 1
- 101001055145 Homo sapiens Interleukin-2 receptor subunit beta Proteins 0.000 description 1
- 101000853002 Homo sapiens Interleukin-25 Proteins 0.000 description 1
- 101001138062 Homo sapiens Leukocyte-associated immunoglobulin-like receptor 1 Proteins 0.000 description 1
- 101001137987 Homo sapiens Lymphocyte activation gene 3 protein Proteins 0.000 description 1
- 101000934338 Homo sapiens Myeloid cell surface antigen CD33 Proteins 0.000 description 1
- 101001128431 Homo sapiens Myeloid-derived growth factor Proteins 0.000 description 1
- 101000836877 Homo sapiens Sialic acid-binding Ig-like lectin 11 Proteins 0.000 description 1
- 101000863882 Homo sapiens Sialic acid-binding Ig-like lectin 7 Proteins 0.000 description 1
- 101000863883 Homo sapiens Sialic acid-binding Ig-like lectin 9 Proteins 0.000 description 1
- 101000831007 Homo sapiens T-cell immunoreceptor with Ig and ITIM domains Proteins 0.000 description 1
- 101000934346 Homo sapiens T-cell surface antigen CD2 Proteins 0.000 description 1
- 101000914484 Homo sapiens T-lymphocyte activation antigen CD80 Proteins 0.000 description 1
- 101000763579 Homo sapiens Toll-like receptor 1 Proteins 0.000 description 1
- 101000831567 Homo sapiens Toll-like receptor 2 Proteins 0.000 description 1
- 101000831496 Homo sapiens Toll-like receptor 3 Proteins 0.000 description 1
- 101000679903 Homo sapiens Tumor necrosis factor receptor superfamily member 25 Proteins 0.000 description 1
- 101000851376 Homo sapiens Tumor necrosis factor receptor superfamily member 8 Proteins 0.000 description 1
- 101000666896 Homo sapiens V-type immunoglobulin domain-containing suppressor of T-cell activation Proteins 0.000 description 1
- 241000713772 Human immunodeficiency virus 1 Species 0.000 description 1
- 206010020751 Hypersensitivity 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
- XDXDZDZNSLXDNA-TZNDIEGXSA-N Idarubicin Chemical compound C1[C@H](N)[C@H](O)[C@H](C)O[C@H]1O[C@@H]1C2=C(O)C(C(=O)C3=CC=CC=C3C3=O)=C3C(O)=C2C[C@@](O)(C(C)=O)C1 XDXDZDZNSLXDNA-TZNDIEGXSA-N 0.000 description 1
- XDXDZDZNSLXDNA-UHFFFAOYSA-N Idarubicin Natural products C1C(N)C(O)C(C)OC1OC1C2=C(O)C(C(=O)C3=CC=CC=C3C3=O)=C3C(O)=C2CC(O)(C(C)=O)C1 XDXDZDZNSLXDNA-UHFFFAOYSA-N 0.000 description 1
- 108010021625 Immunoglobulin Fragments Proteins 0.000 description 1
- 102000008394 Immunoglobulin Fragments Human genes 0.000 description 1
- 108700005091 Immunoglobulin Genes Proteins 0.000 description 1
- 108010016648 Immunophilins Proteins 0.000 description 1
- 102000000521 Immunophilins Human genes 0.000 description 1
- 102100040061 Indoleamine 2,3-dioxygenase 1 Human genes 0.000 description 1
- 102100040062 Indoleamine 2,3-dioxygenase 2 Human genes 0.000 description 1
- 102100021317 Inducible T-cell costimulator Human genes 0.000 description 1
- 101710205775 Inducible T-cell costimulator Proteins 0.000 description 1
- 102000055031 Inhibitor of Apoptosis Proteins Human genes 0.000 description 1
- 102000004877 Insulin Human genes 0.000 description 1
- 108090001061 Insulin Proteins 0.000 description 1
- 108090000723 Insulin-Like Growth Factor I Proteins 0.000 description 1
- 108090001117 Insulin-Like Growth Factor II Proteins 0.000 description 1
- 102100037852 Insulin-like growth factor I Human genes 0.000 description 1
- 102400000022 Insulin-like growth factor II Human genes 0.000 description 1
- 102000006992 Interferon-alpha Human genes 0.000 description 1
- 108010047761 Interferon-alpha Proteins 0.000 description 1
- 102000003996 Interferon-beta Human genes 0.000 description 1
- 108090000467 Interferon-beta Proteins 0.000 description 1
- 102000008070 Interferon-gamma Human genes 0.000 description 1
- 108010074328 Interferon-gamma Proteins 0.000 description 1
- 102000000589 Interleukin-1 Human genes 0.000 description 1
- 108010002352 Interleukin-1 Proteins 0.000 description 1
- 102000051628 Interleukin-1 receptor antagonist Human genes 0.000 description 1
- 108700021006 Interleukin-1 receptor antagonist Proteins 0.000 description 1
- 108090000177 Interleukin-11 Proteins 0.000 description 1
- 108010065805 Interleukin-12 Proteins 0.000 description 1
- 108090000176 Interleukin-13 Proteins 0.000 description 1
- 102000003812 Interleukin-15 Human genes 0.000 description 1
- 108090000172 Interleukin-15 Proteins 0.000 description 1
- 102000049772 Interleukin-16 Human genes 0.000 description 1
- 101800003050 Interleukin-16 Proteins 0.000 description 1
- 108050003558 Interleukin-17 Proteins 0.000 description 1
- 102000013691 Interleukin-17 Human genes 0.000 description 1
- 102000003810 Interleukin-18 Human genes 0.000 description 1
- 108090000171 Interleukin-18 Proteins 0.000 description 1
- 108010002350 Interleukin-2 Proteins 0.000 description 1
- 102100026879 Interleukin-2 receptor subunit beta Human genes 0.000 description 1
- 102100030703 Interleukin-22 Human genes 0.000 description 1
- 108010002386 Interleukin-3 Proteins 0.000 description 1
- 108090000978 Interleukin-4 Proteins 0.000 description 1
- 108010002616 Interleukin-5 Proteins 0.000 description 1
- 108090001005 Interleukin-6 Proteins 0.000 description 1
- 108010002586 Interleukin-7 Proteins 0.000 description 1
- 108090001007 Interleukin-8 Proteins 0.000 description 1
- BKCJZNIZRWYHBN-UHFFFAOYSA-N Isophosphamide mustard Chemical compound ClCCNP(=O)(O)NCCCl BKCJZNIZRWYHBN-UHFFFAOYSA-N 0.000 description 1
- 102100020880 Kit ligand Human genes 0.000 description 1
- 101150030213 Lag3 gene Proteins 0.000 description 1
- 208000031671 Large B-Cell Diffuse Lymphoma Diseases 0.000 description 1
- 102000016267 Leptin Human genes 0.000 description 1
- 108010092277 Leptin Proteins 0.000 description 1
- 102100032352 Leukemia inhibitory factor Human genes 0.000 description 1
- 108090000581 Leukemia inhibitory factor Proteins 0.000 description 1
- 102100020943 Leukocyte-associated immunoglobulin-like receptor 1 Human genes 0.000 description 1
- 239000000232 Lipid Bilayer Substances 0.000 description 1
- 206010025323 Lymphomas Diseases 0.000 description 1
- 102000004083 Lymphotoxin-alpha Human genes 0.000 description 1
- 108090000542 Lymphotoxin-alpha Proteins 0.000 description 1
- 108010061593 Member 14 Tumor Necrosis Factor Receptors Proteins 0.000 description 1
- 102000018697 Membrane Proteins Human genes 0.000 description 1
- 108010052285 Membrane Proteins Proteins 0.000 description 1
- 208000034578 Multiple myelomas Diseases 0.000 description 1
- 241000699666 Mus <mouse, genus> Species 0.000 description 1
- 101100519207 Mus musculus Pdcd1 gene Proteins 0.000 description 1
- 241000204031 Mycoplasma Species 0.000 description 1
- 241000202952 Mycoplasma fermentans Species 0.000 description 1
- 241000202889 Mycoplasma salivarium Species 0.000 description 1
- 102100025243 Myeloid cell surface antigen CD33 Human genes 0.000 description 1
- 102100031789 Myeloid-derived growth factor Human genes 0.000 description 1
- ZZIKIHCNFWXKDY-UHFFFAOYSA-N Myriocin Natural products CCCCCCC(=O)CCCCCCC=CCC(O)C(O)C(N)(CO)C(O)=O ZZIKIHCNFWXKDY-UHFFFAOYSA-N 0.000 description 1
- CRJGESKKUOMBCT-VQTJNVASSA-N N-acetylsphinganine Chemical compound CCCCCCCCCCCCCCC[C@@H](O)[C@H](CO)NC(C)=O CRJGESKKUOMBCT-VQTJNVASSA-N 0.000 description 1
- BLXXJMDCKKHMKV-UHFFFAOYSA-N Nabumetone Chemical compound C1=C(CCC(C)=O)C=CC2=CC(OC)=CC=C21 BLXXJMDCKKHMKV-UHFFFAOYSA-N 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
- 206010028851 Necrosis Diseases 0.000 description 1
- 208000034176 Neoplasms, Germ Cell and Embryonal Diseases 0.000 description 1
- 108010025020 Nerve Growth Factor Proteins 0.000 description 1
- 102000007072 Nerve Growth Factors Human genes 0.000 description 1
- 101100221647 Neurospora crassa (strain ATCC 24698 / 74-OR23-1A / CBS 708.71 / DSM 1257 / FGSC 987) cox-1 gene Proteins 0.000 description 1
- 108090001074 Nucleocapsid Proteins Proteins 0.000 description 1
- SUHOOTKUPISOBE-UHFFFAOYSA-N O-phosphoethanolamine Chemical compound NCCOP(O)(O)=O SUHOOTKUPISOBE-UHFFFAOYSA-N 0.000 description 1
- BZQFBWGGLXLEPQ-UHFFFAOYSA-N O-phosphoryl-L-serine Natural products OC(=O)C(N)COP(O)(O)=O BZQFBWGGLXLEPQ-UHFFFAOYSA-N 0.000 description 1
- 206010030113 Oedema Diseases 0.000 description 1
- 108091034117 Oligonucleotide Proteins 0.000 description 1
- 102000043276 Oncogene Human genes 0.000 description 1
- 108700020796 Oncogene Proteins 0.000 description 1
- 108010036616 P18-I10 peptide Proteins 0.000 description 1
- 101150062589 PTGS1 gene Proteins 0.000 description 1
- 241000711504 Paramyxoviridae Species 0.000 description 1
- 102000003982 Parathyroid hormone Human genes 0.000 description 1
- 108090000445 Parathyroid hormone Proteins 0.000 description 1
- 235000019483 Peanut oil Nutrition 0.000 description 1
- 108010079855 Peptide Aptamers Proteins 0.000 description 1
- 108010003044 Placental Lactogen Proteins 0.000 description 1
- 239000000381 Placental Lactogen Substances 0.000 description 1
- 241001442539 Plasmodium sp. Species 0.000 description 1
- 108010039918 Polylysine Proteins 0.000 description 1
- 101710130420 Probable capsid assembly scaffolding protein Proteins 0.000 description 1
- 101710094000 Programmed cell death 1 ligand 1 Proteins 0.000 description 1
- 108010076181 Proinsulin Proteins 0.000 description 1
- 102000003946 Prolactin Human genes 0.000 description 1
- 108010057464 Prolactin Proteins 0.000 description 1
- 229940123573 Protein synthesis inhibitor Drugs 0.000 description 1
- CZPWVGJYEJSRLH-UHFFFAOYSA-N Pyrimidine Chemical compound C1=CN=CN=C1 CZPWVGJYEJSRLH-UHFFFAOYSA-N 0.000 description 1
- 102100020718 Receptor-type tyrosine-protein kinase FLT3 Human genes 0.000 description 1
- 101710151245 Receptor-type tyrosine-protein kinase FLT3 Proteins 0.000 description 1
- 108090000103 Relaxin Proteins 0.000 description 1
- 102000003743 Relaxin Human genes 0.000 description 1
- 206010039491 Sarcoma Diseases 0.000 description 1
- 101710204410 Scaffold protein Proteins 0.000 description 1
- 102100027125 Sialic acid-binding Ig-like lectin 11 Human genes 0.000 description 1
- 102100029957 Sialic acid-binding Ig-like lectin 5 Human genes 0.000 description 1
- 101710110535 Sialic acid-binding Ig-like lectin 5 Proteins 0.000 description 1
- 102100029946 Sialic acid-binding Ig-like lectin 7 Human genes 0.000 description 1
- 102100029965 Sialic acid-binding Ig-like lectin 9 Human genes 0.000 description 1
- 102000008115 Signaling Lymphocytic Activation Molecule Family Member 1 Human genes 0.000 description 1
- 108010074687 Signaling Lymphocytic Activation Molecule Family Member 1 Proteins 0.000 description 1
- 108010039445 Stem Cell Factor Proteins 0.000 description 1
- 101710172711 Structural protein Proteins 0.000 description 1
- 102100024834 T-cell immunoreceptor with Ig and ITIM domains Human genes 0.000 description 1
- 102100025237 T-cell surface antigen CD2 Human genes 0.000 description 1
- 102100027222 T-lymphocyte activation antigen CD80 Human genes 0.000 description 1
- BPEGJWRSRHCHSN-UHFFFAOYSA-N Temozolomide Chemical compound O=C1N(C)N=NC2=C(C(N)=O)N=CN21 BPEGJWRSRHCHSN-UHFFFAOYSA-N 0.000 description 1
- 102000002938 Thrombospondin Human genes 0.000 description 1
- 108060008245 Thrombospondin Proteins 0.000 description 1
- 102100027010 Toll-like receptor 1 Human genes 0.000 description 1
- 102100024333 Toll-like receptor 2 Human genes 0.000 description 1
- 102100024324 Toll-like receptor 3 Human genes 0.000 description 1
- 102000004887 Transforming Growth Factor beta Human genes 0.000 description 1
- 108090001012 Transforming Growth Factor beta Proteins 0.000 description 1
- 102400001320 Transforming growth factor alpha Human genes 0.000 description 1
- 101800004564 Transforming growth factor alpha Proteins 0.000 description 1
- 102000044209 Tumor Suppressor Genes Human genes 0.000 description 1
- 108700025716 Tumor Suppressor Genes Proteins 0.000 description 1
- 102100028785 Tumor necrosis factor receptor superfamily member 14 Human genes 0.000 description 1
- 102100033728 Tumor necrosis factor receptor superfamily member 18 Human genes 0.000 description 1
- 102100022203 Tumor necrosis factor receptor superfamily member 25 Human genes 0.000 description 1
- 101710165473 Tumor necrosis factor receptor superfamily member 4 Proteins 0.000 description 1
- 102100022153 Tumor necrosis factor receptor superfamily member 4 Human genes 0.000 description 1
- 102100036857 Tumor necrosis factor receptor superfamily member 8 Human genes 0.000 description 1
- 208000006105 Uterine Cervical Neoplasms Diseases 0.000 description 1
- 102100038282 V-type immunoglobulin domain-containing suppressor of T-cell activation Human genes 0.000 description 1
- 108010073929 Vascular Endothelial Growth Factor A Proteins 0.000 description 1
- 102000005789 Vascular Endothelial Growth Factors Human genes 0.000 description 1
- 108010019530 Vascular Endothelial Growth Factors Proteins 0.000 description 1
- 241000251539 Vertebrata <Metazoa> Species 0.000 description 1
- 241001492404 Woodchuck hepatitis virus Species 0.000 description 1
- 229960003697 abatacept Drugs 0.000 description 1
- 230000002159 abnormal effect Effects 0.000 description 1
- 229960001138 acetylsalicylic acid Drugs 0.000 description 1
- 229960004150 aciclovir Drugs 0.000 description 1
- MKUXAQIIEYXACX-UHFFFAOYSA-N aciclovir Chemical compound N1C(N)=NC(=O)C2=C1N(COCCO)C=N2 MKUXAQIIEYXACX-UHFFFAOYSA-N 0.000 description 1
- RJURFGZVJUQBHK-IIXSONLDSA-N actinomycin D Chemical compound C[C@H]1OC(=O)[C@H](C(C)C)N(C)C(=O)CN(C)C(=O)[C@@H]2CCCN2C(=O)[C@@H](C(C)C)NC(=O)[C@H]1NC(=O)C1=C(N)C(=O)C(C)=C2OC(C(C)=CC=C3C(=O)N[C@@H]4C(=O)N[C@@H](C(N5CCC[C@H]5C(=O)N(C)CC(=O)N(C)[C@@H](C(C)C)C(=O)O[C@@H]4C)=O)C(C)C)=C3N=C21 RJURFGZVJUQBHK-IIXSONLDSA-N 0.000 description 1
- 230000004913 activation Effects 0.000 description 1
- 239000004480 active ingredient Substances 0.000 description 1
- 239000000488 activin Substances 0.000 description 1
- 210000005006 adaptive immune system Anatomy 0.000 description 1
- 238000007792 addition Methods 0.000 description 1
- 239000000654 additive Substances 0.000 description 1
- 230000000996 additive effect Effects 0.000 description 1
- 208000009956 adenocarcinoma Diseases 0.000 description 1
- 230000004931 aggregating effect Effects 0.000 description 1
- 125000003295 alanine group Chemical group N[C@@H](C)C(=O)* 0.000 description 1
- 150000001298 alcohols Chemical class 0.000 description 1
- 229960002459 alefacept Drugs 0.000 description 1
- 125000000217 alkyl group Chemical group 0.000 description 1
- 229940100198 alkylating agent Drugs 0.000 description 1
- 239000002168 alkylating agent Substances 0.000 description 1
- 239000013566 allergen Substances 0.000 description 1
- 208000026935 allergic disease Diseases 0.000 description 1
- 230000007815 allergy Effects 0.000 description 1
- 229910000147 aluminium phosphate Inorganic materials 0.000 description 1
- 230000001668 ameliorated effect Effects 0.000 description 1
- QZNJPJDUBTYMRS-UHFFFAOYSA-M amfenac sodium hydrate Chemical compound O.[Na+].NC1=C(CC([O-])=O)C=CC=C1C(=O)C1=CC=CC=C1 QZNJPJDUBTYMRS-UHFFFAOYSA-M 0.000 description 1
- 150000001412 amines Chemical class 0.000 description 1
- 150000001414 amino alcohols Chemical class 0.000 description 1
- 125000003277 amino group Chemical group 0.000 description 1
- 229960004238 anakinra Drugs 0.000 description 1
- 230000000202 analgesic effect Effects 0.000 description 1
- 229940045799 anthracyclines and related substance Drugs 0.000 description 1
- 239000003242 anti bacterial agent Substances 0.000 description 1
- 230000003466 anti-cipated effect Effects 0.000 description 1
- 230000000340 anti-metabolite Effects 0.000 description 1
- 239000000868 anti-mullerian hormone Substances 0.000 description 1
- 230000005875 antibody response Effects 0.000 description 1
- 229940100197 antimetabolite Drugs 0.000 description 1
- 239000002256 antimetabolite Substances 0.000 description 1
- 229940045687 antimetabolites folic acid analogs Drugs 0.000 description 1
- 229940034982 antineoplastic agent Drugs 0.000 description 1
- 229940045719 antineoplastic alkylating agent nitrosoureas Drugs 0.000 description 1
- 239000003972 antineoplastic antibiotic Substances 0.000 description 1
- 229940045985 antineoplastic platinum compound Drugs 0.000 description 1
- 239000003435 antirheumatic agent Substances 0.000 description 1
- 239000003443 antiviral agent Substances 0.000 description 1
- 230000006907 apoptotic process Effects 0.000 description 1
- 238000013459 approach Methods 0.000 description 1
- 239000003125 aqueous solvent Substances 0.000 description 1
- 125000001124 arachidoyl group Chemical group O=C([*])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])[H] 0.000 description 1
- FZCSTZYAHCUGEM-UHFFFAOYSA-N aspergillomarasmine B Natural products OC(=O)CNC(C(O)=O)CNC(C(O)=O)CC(O)=O FZCSTZYAHCUGEM-UHFFFAOYSA-N 0.000 description 1
- 238000003556 assay Methods 0.000 description 1
- 229940092117 atgam Drugs 0.000 description 1
- 230000002238 attenuated effect Effects 0.000 description 1
- AUJRCFUBUPVWSZ-XTZHGVARSA-M auranofin Chemical compound CCP(CC)(CC)=[Au]S[C@@H]1O[C@H](COC(C)=O)[C@@H](OC(C)=O)[C@H](OC(C)=O)[C@H]1OC(C)=O AUJRCFUBUPVWSZ-XTZHGVARSA-M 0.000 description 1
- 229960005207 auranofin Drugs 0.000 description 1
- 229960001799 aurothioglucose Drugs 0.000 description 1
- 230000006472 autoimmune response Effects 0.000 description 1
- 229950002916 avelumab Drugs 0.000 description 1
- 206010064097 avian influenza Diseases 0.000 description 1
- 125000000852 azido group Chemical group *N=[N+]=[N-] 0.000 description 1
- 208000022362 bacterial infectious disease Diseases 0.000 description 1
- 244000052616 bacterial pathogen Species 0.000 description 1
- 229960004669 basiliximab Drugs 0.000 description 1
- OGBUMNBNEWYMNJ-UHFFFAOYSA-N batilol Chemical class CCCCCCCCCCCCCCCCCCOCC(O)CO OGBUMNBNEWYMNJ-UHFFFAOYSA-N 0.000 description 1
- 229960005347 belatacept Drugs 0.000 description 1
- 229960003270 belimumab Drugs 0.000 description 1
- YTKUWDBFDASYHO-UHFFFAOYSA-N bendamustine Chemical compound ClCCN(CCCl)C1=CC=C2N(C)C(CCCC(O)=O)=NC2=C1 YTKUWDBFDASYHO-UHFFFAOYSA-N 0.000 description 1
- 229960002707 bendamustine Drugs 0.000 description 1
- 229950000321 benralizumab Drugs 0.000 description 1
- DRTQHJPVMGBUCF-PSQAKQOGSA-N beta-L-uridine Natural products O[C@H]1[C@@H](O)[C@H](CO)O[C@@H]1N1C(=O)NC(=O)C=C1 DRTQHJPVMGBUCF-PSQAKQOGSA-N 0.000 description 1
- 239000003012 bilayer membrane Substances 0.000 description 1
- 102000023732 binding proteins Human genes 0.000 description 1
- 108091008324 binding proteins Proteins 0.000 description 1
- 230000004071 biological effect Effects 0.000 description 1
- 230000008512 biological response Effects 0.000 description 1
- 201000000053 blastoma Diseases 0.000 description 1
- 229960001561 bleomycin Drugs 0.000 description 1
- OYVAGSVQBOHSSS-UAPAGMARSA-O bleomycin A2 Chemical compound N([C@H](C(=O)N[C@H](C)[C@@H](O)[C@H](C)C(=O)N[C@@H]([C@H](O)C)C(=O)NCCC=1SC=C(N=1)C=1SC=C(N=1)C(=O)NCCC[S+](C)C)[C@@H](O[C@H]1[C@H]([C@@H](O)[C@H](O)[C@H](CO)O1)O[C@@H]1[C@H]([C@@H](OC(N)=O)[C@H](O)[C@@H](CO)O1)O)C=1N=CNC=1)C(=O)C1=NC([C@H](CC(N)=O)NC[C@H](N)C(N)=O)=NC(N)=C1C OYVAGSVQBOHSSS-UAPAGMARSA-O 0.000 description 1
- 108010006025 bovine growth hormone Proteins 0.000 description 1
- 229960003735 brodalumab Drugs 0.000 description 1
- 239000000872 buffer Substances 0.000 description 1
- 229960002092 busulfan Drugs 0.000 description 1
- 229940046731 calcineurin inhibitors Drugs 0.000 description 1
- VSJKWCGYPAHWDS-FQEVSTJZSA-N camptothecin Chemical compound C1=CC=C2C=C(CN3C4=CC5=C(C3=O)COC(=O)[C@]5(O)CC)C4=NC2=C1 VSJKWCGYPAHWDS-FQEVSTJZSA-N 0.000 description 1
- 229940127093 camptothecin Drugs 0.000 description 1
- 229960001838 canakinumab Drugs 0.000 description 1
- 150000001720 carbohydrates Chemical class 0.000 description 1
- 229960004562 carboplatin Drugs 0.000 description 1
- YAYRGNWWLMLWJE-UHFFFAOYSA-L carboplatin Chemical compound O=C1O[Pt](N)(N)OC(=O)C11CCC1 YAYRGNWWLMLWJE-UHFFFAOYSA-L 0.000 description 1
- 239000001768 carboxy methyl cellulose Substances 0.000 description 1
- 235000010948 carboxy methyl cellulose Nutrition 0.000 description 1
- 150000007942 carboxylates Chemical class 0.000 description 1
- 239000008112 carboxymethyl-cellulose Substances 0.000 description 1
- 150000001768 cations Chemical class 0.000 description 1
- 229960000590 celecoxib Drugs 0.000 description 1
- RZEKVGVHFLEQIL-UHFFFAOYSA-N celecoxib Chemical compound C1=CC(C)=CC=C1C1=CC(C(F)(F)F)=NN1C1=CC=C(S(N)(=O)=O)C=C1 RZEKVGVHFLEQIL-UHFFFAOYSA-N 0.000 description 1
- 230000011712 cell development Effects 0.000 description 1
- 230000010261 cell growth Effects 0.000 description 1
- 230000004663 cell proliferation Effects 0.000 description 1
- 210000002421 cell wall Anatomy 0.000 description 1
- 230000001413 cellular effect Effects 0.000 description 1
- 230000033077 cellular process Effects 0.000 description 1
- 229940106189 ceramide Drugs 0.000 description 1
- ZVEQCJWYRWKARO-UHFFFAOYSA-N ceramide Natural products CCCCCCCCCCCCCCC(O)C(=O)NC(CO)C(O)C=CCCC=C(C)CCCCCCCCC ZVEQCJWYRWKARO-UHFFFAOYSA-N 0.000 description 1
- 229960003115 certolizumab pegol Drugs 0.000 description 1
- 201000010881 cervical cancer Diseases 0.000 description 1
- 230000008859 change Effects 0.000 description 1
- 238000006243 chemical reaction Methods 0.000 description 1
- 229960003677 chloroquine Drugs 0.000 description 1
- WHTVZRBIWZFKQO-UHFFFAOYSA-N chloroquine Natural products ClC1=CC=C2C(NC(C)CCCN(CC)CC)=CC=NC2=C1 WHTVZRBIWZFKQO-UHFFFAOYSA-N 0.000 description 1
- 230000002759 chromosomal effect Effects 0.000 description 1
- DQLATGHUWYMOKM-UHFFFAOYSA-L cisplatin Chemical compound N[Pt](N)(Cl)Cl DQLATGHUWYMOKM-UHFFFAOYSA-L 0.000 description 1
- 229960004316 cisplatin Drugs 0.000 description 1
- 239000003184 complementary RNA Substances 0.000 description 1
- 230000000536 complexating effect Effects 0.000 description 1
- 239000000470 constituent Substances 0.000 description 1
- 230000001276 controlling effect Effects 0.000 description 1
- 230000002596 correlated effect Effects 0.000 description 1
- 239000003246 corticosteroid Substances 0.000 description 1
- 230000008878 coupling Effects 0.000 description 1
- 238000010168 coupling process Methods 0.000 description 1
- 238000005859 coupling reaction Methods 0.000 description 1
- 229940111134 coxibs Drugs 0.000 description 1
- 125000004122 cyclic group Chemical group 0.000 description 1
- 239000003260 cyclooxygenase 1 inhibitor Substances 0.000 description 1
- 239000003255 cyclooxygenase 2 inhibitor Substances 0.000 description 1
- 239000000824 cytostatic agent Substances 0.000 description 1
- 230000001085 cytostatic effect Effects 0.000 description 1
- 229960003901 dacarbazine Drugs 0.000 description 1
- 229960002806 daclizumab Drugs 0.000 description 1
- 229960000640 dactinomycin Drugs 0.000 description 1
- 230000006378 damage Effects 0.000 description 1
- STQGQHZAVUOBTE-VGBVRHCVSA-N daunorubicin Chemical compound O([C@H]1C[C@@](O)(CC=2C(O)=C3C(=O)C=4C=CC=C(C=4C(=O)C3=C(O)C=21)OC)C(C)=O)[C@H]1C[C@H](N)[C@H](O)[C@H](C)O1 STQGQHZAVUOBTE-VGBVRHCVSA-N 0.000 description 1
- 229960000975 daunorubicin Drugs 0.000 description 1
- 125000003074 decanoyl group Chemical group [H]C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C(*)=O 0.000 description 1
- 230000003247 decreasing effect Effects 0.000 description 1
- 239000008380 degradant Substances 0.000 description 1
- 239000007857 degradation product Substances 0.000 description 1
- 238000012217 deletion Methods 0.000 description 1
- 230000037430 deletion Effects 0.000 description 1
- CFCUWKMKBJTWLW-UHFFFAOYSA-N deoliosyl-3C-alpha-L-digitoxosyl-MTM Natural products CC=1C(O)=C2C(O)=C3C(=O)C(OC4OC(C)C(O)C(OC5OC(C)C(O)C(OC6OC(C)C(O)C(C)(O)C6)C5)C4)C(C(OC)C(=O)C(O)C(C)O)CC3=CC2=CC=1OC(OC(C)C1O)CC1OC1CC(O)C(O)C(C)O1 CFCUWKMKBJTWLW-UHFFFAOYSA-N 0.000 description 1
- 238000001212 derivatisation Methods 0.000 description 1
- 238000001514 detection method Methods 0.000 description 1
- 229950006137 dexfosfoserine Drugs 0.000 description 1
- NIJJYAXOARWZEE-UHFFFAOYSA-N di-n-propyl-acetic acid Natural products CCCC(C(O)=O)CCC NIJJYAXOARWZEE-UHFFFAOYSA-N 0.000 description 1
- 238000003745 diagnosis Methods 0.000 description 1
- 229910003460 diamond Inorganic materials 0.000 description 1
- 239000010432 diamond Substances 0.000 description 1
- 229960001259 diclofenac Drugs 0.000 description 1
- DCOPUUMXTXDBNB-UHFFFAOYSA-N diclofenac Chemical compound OC(=O)CC1=CC=CC=C1NC1=C(Cl)C=CC=C1Cl DCOPUUMXTXDBNB-UHFFFAOYSA-N 0.000 description 1
- 206010012818 diffuse large B-cell lymphoma Diseases 0.000 description 1
- 229960000616 diflunisal Drugs 0.000 description 1
- HUPFGZXOMWLGNK-UHFFFAOYSA-N diflunisal Chemical compound C1=C(O)C(C(=O)O)=CC(C=2C(=CC(F)=CC=2)F)=C1 HUPFGZXOMWLGNK-UHFFFAOYSA-N 0.000 description 1
- OGQYPPBGSLZBEG-UHFFFAOYSA-N dimethyl(dioctadecyl)azanium Chemical compound CCCCCCCCCCCCCCCCCC[N+](C)(C)CCCCCCCCCCCCCCCCCC OGQYPPBGSLZBEG-UHFFFAOYSA-N 0.000 description 1
- 230000005750 disease progression Effects 0.000 description 1
- ZGSPNIOCEDOHGS-UHFFFAOYSA-L disodium [3-[2,3-di(octadeca-9,12-dienoyloxy)propoxy-oxidophosphoryl]oxy-2-hydroxypropyl] 2,3-di(octadeca-9,12-dienoyloxy)propyl phosphate Chemical class [Na+].[Na+].CCCCCC=CCC=CCCCCCCCC(=O)OCC(OC(=O)CCCCCCCC=CCC=CCCCCC)COP([O-])(=O)OCC(O)COP([O-])(=O)OCC(OC(=O)CCCCCCCC=CCC=CCCCCC)COC(=O)CCCCCCCC=CCC=CCCCCC ZGSPNIOCEDOHGS-UHFFFAOYSA-L 0.000 description 1
- VSJKWCGYPAHWDS-UHFFFAOYSA-N dl-camptothecin Natural products C1=CC=C2C=C(CN3C4=CC5=C(C3=O)COC(=O)C5(O)CC)C4=NC2=C1 VSJKWCGYPAHWDS-UHFFFAOYSA-N 0.000 description 1
- 229960004679 doxorubicin Drugs 0.000 description 1
- 229950003468 dupilumab Drugs 0.000 description 1
- 229950009791 durvalumab Drugs 0.000 description 1
- 229960002224 eculizumab Drugs 0.000 description 1
- 229960000284 efalizumab Drugs 0.000 description 1
- 239000012636 effector Substances 0.000 description 1
- 235000013601 eggs Nutrition 0.000 description 1
- 230000009881 electrostatic interaction Effects 0.000 description 1
- 201000008184 embryoma Diseases 0.000 description 1
- 210000001671 embryonic stem cell Anatomy 0.000 description 1
- 230000002255 enzymatic effect Effects 0.000 description 1
- 229950006370 epacadostat Drugs 0.000 description 1
- 229960001904 epirubicin Drugs 0.000 description 1
- 125000001901 erucoyl group Chemical group O=C([*])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])/C([H])=C([H])\C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])[H] 0.000 description 1
- 229960000403 etanercept Drugs 0.000 description 1
- 150000002170 ethers Chemical class 0.000 description 1
- 229960005293 etodolac Drugs 0.000 description 1
- XFBVBWWRPKNWHW-UHFFFAOYSA-N etodolac Chemical compound C1COC(CC)(CC(O)=O)C2=N[C]3C(CC)=CC=CC3=C21 XFBVBWWRPKNWHW-UHFFFAOYSA-N 0.000 description 1
- VJJPUSNTGOMMGY-MRVIYFEKSA-N etoposide Chemical compound COC1=C(O)C(OC)=CC([C@@H]2C3=CC=4OCOC=4C=C3[C@@H](O[C@H]3[C@@H]([C@@H](O)[C@@H]4O[C@H](C)OC[C@H]4O3)O)[C@@H]3[C@@H]2C(OC3)=O)=C1 VJJPUSNTGOMMGY-MRVIYFEKSA-N 0.000 description 1
- 229960005420 etoposide Drugs 0.000 description 1
- 231100000776 exotoxin Toxicity 0.000 description 1
- 239000002095 exotoxin Substances 0.000 description 1
- 230000008622 extracellular signaling Effects 0.000 description 1
- 150000002190 fatty acyls Chemical class 0.000 description 1
- 229960001419 fenoprofen Drugs 0.000 description 1
- 229940126864 fibroblast growth factor Drugs 0.000 description 1
- 239000012847 fine chemical Substances 0.000 description 1
- GNBHRKFJIUUOQI-UHFFFAOYSA-N fluorescein Chemical compound O1C(=O)C2=CC=CC=C2C21C1=CC=C(O)C=C1OC1=CC(O)=CC=C21 GNBHRKFJIUUOQI-UHFFFAOYSA-N 0.000 description 1
- 125000001153 fluoro group Chemical group F* 0.000 description 1
- IJJVMEJXYNJXOJ-UHFFFAOYSA-N fluquinconazole Chemical compound C=1C=C(Cl)C=C(Cl)C=1N1C(=O)C2=CC(F)=CC=C2N=C1N1C=NC=N1 IJJVMEJXYNJXOJ-UHFFFAOYSA-N 0.000 description 1
- SYTBZMRGLBWNTM-UHFFFAOYSA-N flurbiprofen Chemical compound FC1=CC(C(C(O)=O)C)=CC=C1C1=CC=CC=C1 SYTBZMRGLBWNTM-UHFFFAOYSA-N 0.000 description 1
- 150000002224 folic acids Chemical class 0.000 description 1
- 239000012537 formulation buffer Substances 0.000 description 1
- 230000004927 fusion Effects 0.000 description 1
- IRSCQMHQWWYFCW-UHFFFAOYSA-N ganciclovir Chemical compound O=C1NC(N)=NC2=C1N=CN2COC(CO)CO IRSCQMHQWWYFCW-UHFFFAOYSA-N 0.000 description 1
- 229960002963 ganciclovir Drugs 0.000 description 1
- 150000002270 gangliosides Chemical class 0.000 description 1
- 239000007789 gas Substances 0.000 description 1
- SDUQYLNIPVEERB-QPPQHZFASA-N gemcitabine Chemical compound O=C1N=C(N)C=CN1[C@H]1C(F)(F)[C@H](O)[C@@H](CO)O1 SDUQYLNIPVEERB-QPPQHZFASA-N 0.000 description 1
- 229960005277 gemcitabine Drugs 0.000 description 1
- 230000002068 genetic effect Effects 0.000 description 1
- 102000054766 genetic haplotypes Human genes 0.000 description 1
- 239000003862 glucocorticoid Substances 0.000 description 1
- AWUCVROLDVIAJX-UHFFFAOYSA-N glycerol 1-phosphate Chemical compound OCC(O)COP(O)(O)=O AWUCVROLDVIAJX-UHFFFAOYSA-N 0.000 description 1
- 150000002327 glycerophospholipids Chemical class 0.000 description 1
- 125000003976 glyceryl group Chemical group [H]C([*])([H])C(O[H])([H])C(O[H])([H])[H] 0.000 description 1
- 229940015045 gold sodium thiomalate Drugs 0.000 description 1
- 229960001743 golimumab Drugs 0.000 description 1
- 239000002622 gonadotropin Substances 0.000 description 1
- 230000012010 growth Effects 0.000 description 1
- 229950010864 guselkumab Drugs 0.000 description 1
- 201000010536 head and neck cancer Diseases 0.000 description 1
- 208000014829 head and neck neoplasm Diseases 0.000 description 1
- 230000002440 hepatic effect Effects 0.000 description 1
- 210000004408 hybridoma Anatomy 0.000 description 1
- 229960000890 hydrocortisone Drugs 0.000 description 1
- 230000007062 hydrolysis Effects 0.000 description 1
- 238000006460 hydrolysis reaction Methods 0.000 description 1
- 125000002887 hydroxy group Chemical group [H]O* 0.000 description 1
- XXSMGPRMXLTPCZ-UHFFFAOYSA-N hydroxychloroquine Chemical compound ClC1=CC=C2C(NC(C)CCCN(CCO)CC)=CC=NC2=C1 XXSMGPRMXLTPCZ-UHFFFAOYSA-N 0.000 description 1
- 229960004171 hydroxychloroquine Drugs 0.000 description 1
- 229960001680 ibuprofen Drugs 0.000 description 1
- 229960000908 idarubicin Drugs 0.000 description 1
- 229960001101 ifosfamide Drugs 0.000 description 1
- HOMGKSMUEGBAAB-UHFFFAOYSA-N ifosfamide Chemical compound ClCCNP1(=O)OCCCN1CCCl HOMGKSMUEGBAAB-UHFFFAOYSA-N 0.000 description 1
- 229960002751 imiquimod Drugs 0.000 description 1
- DOUYETYNHWVLEO-UHFFFAOYSA-N imiquimod Chemical compound C1=CC=CC2=C3N(CC(C)C)C=NC3=C(N)N=C21 DOUYETYNHWVLEO-UHFFFAOYSA-N 0.000 description 1
- 102000027596 immune receptors Human genes 0.000 description 1
- 108091008915 immune receptors Proteins 0.000 description 1
- 229960003444 immunosuppressant agent Drugs 0.000 description 1
- 230000001861 immunosuppressant effect Effects 0.000 description 1
- 238000000338 in vitro Methods 0.000 description 1
- 229960000905 indomethacin Drugs 0.000 description 1
- 239000000893 inhibin Substances 0.000 description 1
- 230000002401 inhibitory effect Effects 0.000 description 1
- ZPNFWUPYTFPOJU-LPYSRVMUSA-N iniprol Chemical compound C([C@H]1C(=O)NCC(=O)NCC(=O)N[C@H]2CSSC[C@H]3C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](C)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@H](C(N[C@H](C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC=4C=CC(O)=CC=4)C(=O)N[C@@H](CC=4C=CC=CC=4)C(=O)N[C@@H](CC=4C=CC(O)=CC=4)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](C)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](C)C(=O)NCC(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CSSC[C@H](NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](C)NC(=O)[C@H](CO)NC(=O)[C@H](CCCCN)NC(=O)[C@H](CC=4C=CC=CC=4)NC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CCCCN)NC(=O)[C@H](C)NC(=O)[C@H](CCCNC(N)=N)NC2=O)C(=O)N[C@@H](CCSC)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CSSC[C@H](NC(=O)[C@H](CC=2C=CC=CC=2)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H]2N(CCC2)C(=O)[C@@H](N)CCCNC(N)=N)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCC(O)=O)C(=O)N2[C@@H](CCC2)C(=O)N2[C@@H](CCC2)C(=O)N[C@@H](CC=2C=CC(O)=CC=2)C(=O)N[C@@H]([C@@H](C)O)C(=O)NCC(=O)N2[C@@H](CCC2)C(=O)N3)C(=O)NCC(=O)NCC(=O)N[C@@H](C)C(O)=O)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@H](C(=O)N[C@@H](CC=2C=CC=CC=2)C(=O)N[C@H](C(=O)N1)C(C)C)[C@@H](C)O)[C@@H](C)CC)=O)[C@@H](C)CC)C1=CC=C(O)C=C1 ZPNFWUPYTFPOJU-LPYSRVMUSA-N 0.000 description 1
- 238000002347 injection Methods 0.000 description 1
- 239000007924 injection Substances 0.000 description 1
- 230000015788 innate immune response Effects 0.000 description 1
- 229940125396 insulin Drugs 0.000 description 1
- 102000006495 integrins Human genes 0.000 description 1
- 108010044426 integrins Proteins 0.000 description 1
- 230000010468 interferon response Effects 0.000 description 1
- 229940046732 interleukin inhibitors Drugs 0.000 description 1
- 108010074108 interleukin-21 Proteins 0.000 description 1
- 230000003834 intracellular effect Effects 0.000 description 1
- 150000002500 ions Chemical class 0.000 description 1
- 229960005386 ipilimumab Drugs 0.000 description 1
- 229960004768 irinotecan Drugs 0.000 description 1
- UWKQSNNFCGGAFS-XIFFEERXSA-N irinotecan Chemical compound C1=C2C(CC)=C3CN(C(C4=C([C@@](C(=O)OC4)(O)CC)C=4)=O)C=4C3=NC2=CC=C1OC(=O)N(CC1)CCC1N1CCCCC1 UWKQSNNFCGGAFS-XIFFEERXSA-N 0.000 description 1
- 230000001788 irregular Effects 0.000 description 1
- 229960005435 ixekizumab Drugs 0.000 description 1
- DKYWVDODHFEZIM-UHFFFAOYSA-N ketoprofen Chemical compound OC(=O)C(C)C1=CC=CC(C(=O)C=2C=CC=CC=2)=C1 DKYWVDODHFEZIM-UHFFFAOYSA-N 0.000 description 1
- 229960000991 ketoprofen Drugs 0.000 description 1
- 229960004752 ketorolac Drugs 0.000 description 1
- OZWKMVRBQXNZKK-UHFFFAOYSA-N ketorolac Chemical compound OC(=O)C1CCN2C1=CC=C2C(=O)C1=CC=CC=C1 OZWKMVRBQXNZKK-UHFFFAOYSA-N 0.000 description 1
- 125000000400 lauroyl group Chemical group O=C([*])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])[H] 0.000 description 1
- 229960000681 leflunomide Drugs 0.000 description 1
- VHOGYURTWQBHIL-UHFFFAOYSA-N leflunomide Chemical compound O1N=CC(C(=O)NC=2C=CC(=CC=2)C(F)(F)F)=C1C VHOGYURTWQBHIL-UHFFFAOYSA-N 0.000 description 1
- 231100000518 lethal Toxicity 0.000 description 1
- 230000001665 lethal effect Effects 0.000 description 1
- 125000002669 linoleoyl group Chemical group O=C([*])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])/C([H])=C([H])\C([H])([H])/C([H])=C([H])\C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])[H] 0.000 description 1
- 229920006008 lipopolysaccharide Polymers 0.000 description 1
- 229920002521 macromolecule Polymers 0.000 description 1
- 108091005446 macrophage receptors Proteins 0.000 description 1
- 108010051618 macrophage stimulatory lipopeptide 2 Proteins 0.000 description 1
- 230000036210 malignancy Effects 0.000 description 1
- 238000001840 matrix-assisted laser desorption--ionisation time-of-flight mass spectrometry Methods 0.000 description 1
- 238000005259 measurement Methods 0.000 description 1
- 230000007246 mechanism Effects 0.000 description 1
- SGDBTWWWUNNDEQ-LBPRGKRZSA-N melphalan Chemical compound OC(=O)[C@@H](N)CC1=CC=C(N(CCCl)CCCl)C=C1 SGDBTWWWUNNDEQ-LBPRGKRZSA-N 0.000 description 1
- 229960001924 melphalan Drugs 0.000 description 1
- 229960005108 mepolizumab Drugs 0.000 description 1
- GLVAUDGFNGKCSF-UHFFFAOYSA-N mercaptopurine Chemical compound S=C1NC=NC2=C1NC=N2 GLVAUDGFNGKCSF-UHFFFAOYSA-N 0.000 description 1
- 229960001428 mercaptopurine Drugs 0.000 description 1
- 230000001394 metastastic effect Effects 0.000 description 1
- 208000037819 metastatic cancer Diseases 0.000 description 1
- 208000011575 metastatic malignant neoplasm Diseases 0.000 description 1
- HPNSFSBZBAHARI-UHFFFAOYSA-N micophenolic acid Natural products OC1=C(CC=C(C)CCC(O)=O)C(OC)=C(C)C2=C1C(=O)OC2 HPNSFSBZBAHARI-UHFFFAOYSA-N 0.000 description 1
- 244000005700 microbiome Species 0.000 description 1
- CFCUWKMKBJTWLW-BKHRDMLASA-N mithramycin Chemical compound O([C@@H]1C[C@@H](O[C@H](C)[C@H]1O)OC=1C=C2C=C3C[C@H]([C@@H](C(=O)C3=C(O)C2=C(O)C=1C)O[C@@H]1O[C@H](C)[C@@H](O)[C@H](O[C@@H]2O[C@H](C)[C@H](O)[C@H](O[C@@H]3O[C@H](C)[C@@H](O)[C@@](C)(O)C3)C2)C1)[C@H](OC)C(=O)[C@@H](O)[C@@H](C)O)[C@H]1C[C@@H](O)[C@H](O)[C@@H](C)O1 CFCUWKMKBJTWLW-BKHRDMLASA-N 0.000 description 1
- KKZJGLLVHKMTCM-UHFFFAOYSA-N mitoxantrone Chemical compound O=C1C2=C(O)C=CC(O)=C2C(=O)C2=C1C(NCCNCCO)=CC=C2NCCNCCO KKZJGLLVHKMTCM-UHFFFAOYSA-N 0.000 description 1
- 229960001156 mitoxantrone Drugs 0.000 description 1
- 239000003607 modifier Substances 0.000 description 1
- 229960003816 muromonab-cd3 Drugs 0.000 description 1
- 238000002703 mutagenesis Methods 0.000 description 1
- 231100000350 mutagenesis Toxicity 0.000 description 1
- 229960004866 mycophenolate mofetil Drugs 0.000 description 1
- RTGDFNSFWBGLEC-SYZQJQIISA-N mycophenolate mofetil Chemical compound COC1=C(C)C=2COC(=O)C=2C(O)=C1C\C=C(/C)CCC(=O)OCCN1CCOCC1 RTGDFNSFWBGLEC-SYZQJQIISA-N 0.000 description 1
- 229960000951 mycophenolic acid Drugs 0.000 description 1
- HPNSFSBZBAHARI-RUDMXATFSA-N mycophenolic acid Chemical compound OC1=C(C\C=C(/C)CCC(O)=O)C(OC)=C(C)C2=C1C(=O)OC2 HPNSFSBZBAHARI-RUDMXATFSA-N 0.000 description 1
- 201000000050 myeloid neoplasm Diseases 0.000 description 1
- ZZIKIHCNFWXKDY-GNTQXERDSA-N myriocin Chemical compound CCCCCCC(=O)CCCCCC\C=C\C[C@@H](O)[C@H](O)[C@@](N)(CO)C(O)=O ZZIKIHCNFWXKDY-GNTQXERDSA-N 0.000 description 1
- 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 1
- 229960004270 nabumetone Drugs 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
- 230000017074 necrotic cell death Effects 0.000 description 1
- 230000006654 negative regulation of apoptotic process Effects 0.000 description 1
- VVGIYYKRAMHVLU-UHFFFAOYSA-N newbouldiamide Natural products CCCCCCCCCCCCCCCCCCCC(O)C(O)C(O)C(CO)NC(=O)CCCCCCCCCCCCCCCCC VVGIYYKRAMHVLU-UHFFFAOYSA-N 0.000 description 1
- 229910052759 nickel Inorganic materials 0.000 description 1
- QJGQUHMNIGDVPM-UHFFFAOYSA-N nitrogen group Chemical group [N] QJGQUHMNIGDVPM-UHFFFAOYSA-N 0.000 description 1
- 229960003301 nivolumab Drugs 0.000 description 1
- 230000000683 nonmetastatic effect Effects 0.000 description 1
- 239000002777 nucleoside Substances 0.000 description 1
- 125000003835 nucleoside group Chemical group 0.000 description 1
- ZQPPMHVWECSIRJ-KTKRTIGZSA-M oleate Chemical compound CCCCCCCC\C=C/CCCCCCCC([O-])=O ZQPPMHVWECSIRJ-KTKRTIGZSA-M 0.000 description 1
- 125000002811 oleoyl group Chemical group O=C([*])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])/C([H])=C([H])\C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])[H] 0.000 description 1
- 229960000470 omalizumab Drugs 0.000 description 1
- 238000011275 oncology therapy Methods 0.000 description 1
- 230000003287 optical effect Effects 0.000 description 1
- 230000002138 osteoinductive effect Effects 0.000 description 1
- DWAFYCQODLXJNR-BNTLRKBRSA-L oxaliplatin Chemical compound O1C(=O)C(=O)O[Pt]11N[C@@H]2CCCC[C@H]2N1 DWAFYCQODLXJNR-BNTLRKBRSA-L 0.000 description 1
- 229960001756 oxaliplatin Drugs 0.000 description 1
- OFPXSFXSNFPTHF-UHFFFAOYSA-N oxaprozin Chemical compound O1C(CCC(=O)O)=NC(C=2C=CC=CC=2)=C1C1=CC=CC=C1 OFPXSFXSNFPTHF-UHFFFAOYSA-N 0.000 description 1
- 229960002739 oxaprozin Drugs 0.000 description 1
- 239000005022 packaging material Substances 0.000 description 1
- 229950000755 palifosfamide Drugs 0.000 description 1
- 239000000199 parathyroid hormone Substances 0.000 description 1
- 229960001319 parathyroid hormone Drugs 0.000 description 1
- 230000037361 pathway Effects 0.000 description 1
- 102000007863 pattern recognition receptors Human genes 0.000 description 1
- 108010089193 pattern recognition receptors Proteins 0.000 description 1
- 239000000312 peanut oil Substances 0.000 description 1
- 229960002621 pembrolizumab Drugs 0.000 description 1
- 229960001639 penicillamine Drugs 0.000 description 1
- 238000002823 phage display Methods 0.000 description 1
- 239000010452 phosphate Substances 0.000 description 1
- 235000021317 phosphate Nutrition 0.000 description 1
- 150000004713 phosphodiesters Chemical class 0.000 description 1
- 150000003013 phosphoric acid derivatives Chemical class 0.000 description 1
- BZQFBWGGLXLEPQ-REOHCLBHSA-N phosphoserine Chemical compound OC(=O)[C@@H](N)COP(O)(O)=O BZQFBWGGLXLEPQ-REOHCLBHSA-N 0.000 description 1
- 229950010773 pidilizumab Drugs 0.000 description 1
- 229960005330 pimecrolimus Drugs 0.000 description 1
- KASDHRXLYQOAKZ-ZPSXYTITSA-N pimecrolimus Chemical compound C/C([C@H]1OC(=O)[C@@H]2CCCCN2C(=O)C(=O)[C@]2(O)O[C@@H]([C@H](C[C@H]2C)OC)[C@@H](OC)C[C@@H](C)C/C(C)=C/[C@H](C(C[C@H](O)[C@H]1C)=O)CC)=C\[C@@H]1CC[C@@H](Cl)[C@H](OC)C1 KASDHRXLYQOAKZ-ZPSXYTITSA-N 0.000 description 1
- QYSPLQLAKJAUJT-UHFFFAOYSA-N piroxicam Chemical compound OC=1C2=CC=CC=C2S(=O)(=O)N(C)C=1C(=O)NC1=CC=CC=N1 QYSPLQLAKJAUJT-UHFFFAOYSA-N 0.000 description 1
- 229960002702 piroxicam Drugs 0.000 description 1
- PEZPMAYDXJQYRV-UHFFFAOYSA-N pixantrone Chemical compound O=C1C2=CN=CC=C2C(=O)C2=C1C(NCCN)=CC=C2NCCN PEZPMAYDXJQYRV-UHFFFAOYSA-N 0.000 description 1
- 229960004403 pixantrone Drugs 0.000 description 1
- 210000004180 plasmocyte Anatomy 0.000 description 1
- 150000003058 platinum compounds Chemical class 0.000 description 1
- 229960003171 plicamycin Drugs 0.000 description 1
- 229920001223 polyethylene glycol Polymers 0.000 description 1
- 229940115272 polyinosinic:polycytidylic acid Drugs 0.000 description 1
- 229920000656 polylysine Polymers 0.000 description 1
- 229960000688 pomalidomide Drugs 0.000 description 1
- UVSMNLNDYGZFPF-UHFFFAOYSA-N pomalidomide Chemical compound O=C1C=2C(N)=CC=CC=2C(=O)N1C1CCC(=O)NC1=O UVSMNLNDYGZFPF-UHFFFAOYSA-N 0.000 description 1
- 239000011148 porous material Substances 0.000 description 1
- 230000032361 posttranscriptional gene silencing Effects 0.000 description 1
- 238000001556 precipitation Methods 0.000 description 1
- 230000003449 preventive effect Effects 0.000 description 1
- 125000002924 primary amino group Chemical group [H]N([H])* 0.000 description 1
- 230000007112 pro inflammatory response Effects 0.000 description 1
- CPTBDICYNRMXFX-UHFFFAOYSA-N procarbazine Chemical compound CNNCC1=CC=C(C(=O)NC(C)C)C=C1 CPTBDICYNRMXFX-UHFFFAOYSA-N 0.000 description 1
- 229960000624 procarbazine Drugs 0.000 description 1
- 238000012545 processing Methods 0.000 description 1
- 229940097325 prolactin Drugs 0.000 description 1
- 230000000069 prophylactic effect Effects 0.000 description 1
- 238000011321 prophylaxis Methods 0.000 description 1
- 108010087851 prorelaxin Proteins 0.000 description 1
- 150000003180 prostaglandins Chemical class 0.000 description 1
- 230000001681 protective effect Effects 0.000 description 1
- 230000020978 protein processing Effects 0.000 description 1
- 239000000007 protein synthesis inhibitor Substances 0.000 description 1
- 244000079416 protozoan pathogen Species 0.000 description 1
- 150000003212 purines Chemical class 0.000 description 1
- 150000003230 pyrimidines Chemical class 0.000 description 1
- 238000011002 quantification Methods 0.000 description 1
- 229940121896 radiopharmaceutical Drugs 0.000 description 1
- 239000012217 radiopharmaceutical Substances 0.000 description 1
- 230000002799 radiopharmaceutical effect Effects 0.000 description 1
- 230000006798 recombination Effects 0.000 description 1
- 210000003289 regulatory T cell Anatomy 0.000 description 1
- 230000001105 regulatory effect Effects 0.000 description 1
- 229960003254 reslizumab Drugs 0.000 description 1
- 238000012552 review Methods 0.000 description 1
- 229960001886 rilonacept Drugs 0.000 description 1
- 108010046141 rilonacept Proteins 0.000 description 1
- 229960000953 salsalate Drugs 0.000 description 1
- 229950006348 sarilumab Drugs 0.000 description 1
- 238000012216 screening Methods 0.000 description 1
- 230000003248 secreting effect Effects 0.000 description 1
- 229960004540 secukinumab Drugs 0.000 description 1
- 229940046729 selective immunosuppressants Drugs 0.000 description 1
- 108091006024 signal transducing proteins Proteins 0.000 description 1
- 102000034285 signal transducing proteins Human genes 0.000 description 1
- 229960003323 siltuximab Drugs 0.000 description 1
- 238000000235 small-angle X-ray scattering Methods 0.000 description 1
- AGHLUVOCTHWMJV-UHFFFAOYSA-J sodium;gold(3+);2-sulfanylbutanedioate Chemical compound [Na+].[Au+3].[O-]C(=O)CC(S)C([O-])=O.[O-]C(=O)CC(S)C([O-])=O AGHLUVOCTHWMJV-UHFFFAOYSA-J 0.000 description 1
- 239000007790 solid phase Substances 0.000 description 1
- 230000007928 solubilization Effects 0.000 description 1
- 238000005063 solubilization Methods 0.000 description 1
- 230000037439 somatic mutation Effects 0.000 description 1
- 239000001593 sorbitan monooleate Substances 0.000 description 1
- 235000011069 sorbitan monooleate Nutrition 0.000 description 1
- 229940035049 sorbitan monooleate Drugs 0.000 description 1
- 235000012424 soybean oil Nutrition 0.000 description 1
- 239000003549 soybean oil Substances 0.000 description 1
- 125000006850 spacer group Chemical group 0.000 description 1
- 150000003408 sphingolipids Chemical class 0.000 description 1
- 210000003046 sporozoite Anatomy 0.000 description 1
- 230000006641 stabilisation Effects 0.000 description 1
- 238000010561 standard procedure Methods 0.000 description 1
- 210000000130 stem cell Anatomy 0.000 description 1
- 239000008223 sterile water Substances 0.000 description 1
- 230000000638 stimulation Effects 0.000 description 1
- 238000003756 stirring Methods 0.000 description 1
- 238000010254 subcutaneous injection Methods 0.000 description 1
- 239000007929 subcutaneous injection Substances 0.000 description 1
- 230000002311 subsequent effect Effects 0.000 description 1
- 238000006467 substitution reaction Methods 0.000 description 1
- 150000003871 sulfonates Chemical class 0.000 description 1
- 150000003467 sulfuric acid derivatives Chemical class 0.000 description 1
- MLKXDPUZXIRXEP-MFOYZWKCSA-N sulindac Chemical compound CC1=C(CC(O)=O)C2=CC(F)=CC=C2\C1=C/C1=CC=C(S(C)=O)C=C1 MLKXDPUZXIRXEP-MFOYZWKCSA-N 0.000 description 1
- 229960000894 sulindac Drugs 0.000 description 1
- 230000004083 survival effect Effects 0.000 description 1
- 230000002195 synergetic effect Effects 0.000 description 1
- 238000003786 synthesis reaction Methods 0.000 description 1
- 229960004964 temozolomide Drugs 0.000 description 1
- NRUKOCRGYNPUPR-QBPJDGROSA-N teniposide Chemical compound COC1=C(O)C(OC)=CC([C@@H]2C3=CC=4OCOC=4C=C3[C@@H](O[C@H]3[C@@H]([C@@H](O)[C@@H]4O[C@@H](OC[C@H]4O3)C=3SC=CC=3)O)[C@@H]3[C@@H]2C(OC3)=O)=C1 NRUKOCRGYNPUPR-QBPJDGROSA-N 0.000 description 1
- 229960001278 teniposide Drugs 0.000 description 1
- 229960000331 teriflunomide Drugs 0.000 description 1
- UTNUDOFZCWSZMS-YFHOEESVSA-N teriflunomide Chemical compound C\C(O)=C(/C#N)C(=O)NC1=CC=C(C(F)(F)F)C=C1 UTNUDOFZCWSZMS-YFHOEESVSA-N 0.000 description 1
- ZRKFYGHZFMAOKI-QMGMOQQFSA-N tgfbeta Chemical compound C([C@H](NC(=O)[C@H](C(C)C)NC(=O)CNC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H]([C@@H](C)O)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H]([C@@H](C)O)NC(=O)[C@H](CC(C)C)NC(=O)CNC(=O)[C@H](C)NC(=O)[C@H](CO)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@@H](NC(=O)[C@H](C)NC(=O)[C@H](C)NC(=O)[C@@H](NC(=O)[C@H](CC(C)C)NC(=O)[C@@H](N)CCSC)C(C)C)[C@@H](C)CC)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](C)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](C)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](C)C(=O)N[C@@H](CC(C)C)C(=O)N1[C@@H](CCC1)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CO)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC(C)C)C(O)=O)C1=CC=C(O)C=C1 ZRKFYGHZFMAOKI-QMGMOQQFSA-N 0.000 description 1
- 229960003433 thalidomide Drugs 0.000 description 1
- 230000004797 therapeutic response Effects 0.000 description 1
- 150000007970 thio esters Chemical class 0.000 description 1
- RYYWUUFWQRZTIU-UHFFFAOYSA-K thiophosphate Chemical compound [O-]P([O-])([O-])=S RYYWUUFWQRZTIU-UHFFFAOYSA-K 0.000 description 1
- 229940107955 thymoglobulin Drugs 0.000 description 1
- 229940034208 thyroxine Drugs 0.000 description 1
- XUIIKFGFIJCVMT-UHFFFAOYSA-N thyroxine-binding globulin Natural products IC1=CC(CC([NH3+])C([O-])=O)=CC(I)=C1OC1=CC(I)=C(O)C(I)=C1 XUIIKFGFIJCVMT-UHFFFAOYSA-N 0.000 description 1
- 229960003087 tioguanine Drugs 0.000 description 1
- 229960003989 tocilizumab Drugs 0.000 description 1
- 229960001017 tolmetin Drugs 0.000 description 1
- UPSPUYADGBWSHF-UHFFFAOYSA-N tolmetin Chemical compound C1=CC(C)=CC=C1C(=O)C1=CC=C(CC(O)=O)N1C UPSPUYADGBWSHF-UHFFFAOYSA-N 0.000 description 1
- UCFGDBYHRUNTLO-QHCPKHFHSA-N topotecan Chemical compound C1=C(O)C(CN(C)C)=C2C=C(CN3C4=CC5=C(C3=O)COC(=O)[C@]5(O)CC)C4=NC2=C1 UCFGDBYHRUNTLO-QHCPKHFHSA-N 0.000 description 1
- 229960000303 topotecan Drugs 0.000 description 1
- PKVRCIRHQMSYJX-AIFWHQITSA-N trabectedin Chemical compound C([C@@]1(C(OC2)=O)NCCC3=C1C=C(C(=C3)O)OC)S[C@@H]1C3=C(OC(C)=O)C(C)=C4OCOC4=C3[C@H]2N2[C@@H](O)[C@H](CC=3C4=C(O)C(OC)=C(C)C=3)N(C)[C@H]4[C@@H]21 PKVRCIRHQMSYJX-AIFWHQITSA-N 0.000 description 1
- 238000013518 transcription Methods 0.000 description 1
- 230000035897 transcription Effects 0.000 description 1
- 238000010361 transduction Methods 0.000 description 1
- 230000026683 transduction Effects 0.000 description 1
- 238000003151 transfection method Methods 0.000 description 1
- 230000014616 translation Effects 0.000 description 1
- 238000013519 translation Methods 0.000 description 1
- 229950007217 tremelimumab Drugs 0.000 description 1
- 150000003626 triacylglycerols Chemical class 0.000 description 1
- 230000001960 triggered effect Effects 0.000 description 1
- 210000004881 tumor cell Anatomy 0.000 description 1
- 241001529453 unidentified herpesvirus Species 0.000 description 1
- 239000002691 unilamellar liposome Substances 0.000 description 1
- 229930195735 unsaturated hydrocarbon Natural products 0.000 description 1
- DRTQHJPVMGBUCF-UHFFFAOYSA-N uracil arabinoside Natural products OC1C(O)C(CO)OC1N1C(=O)NC(=O)C=C1 DRTQHJPVMGBUCF-UHFFFAOYSA-N 0.000 description 1
- 229960001055 uracil mustard Drugs 0.000 description 1
- 229940045145 uridine Drugs 0.000 description 1
- 229960003824 ustekinumab Drugs 0.000 description 1
- MSRILKIQRXUYCT-UHFFFAOYSA-M valproate semisodium Chemical compound [Na+].CCCC(C(O)=O)CCC.CCCC(C([O-])=O)CCC MSRILKIQRXUYCT-UHFFFAOYSA-M 0.000 description 1
- 229960000604 valproic acid Drugs 0.000 description 1
- 229960004914 vedolizumab Drugs 0.000 description 1
- 235000013311 vegetables Nutrition 0.000 description 1
- 244000052613 viral pathogen Species 0.000 description 1
- 210000002845 virion Anatomy 0.000 description 1
- 230000000007 visual effect Effects 0.000 description 1
- WAEXFXRVDQXREF-UHFFFAOYSA-N vorinostat Chemical compound ONC(=O)CCCCCCC(=O)NC1=CC=CC=C1 WAEXFXRVDQXREF-UHFFFAOYSA-N 0.000 description 1
- 229960000237 vorinostat Drugs 0.000 description 1
- 238000010792 warming Methods 0.000 description 1
- 229940004212 yondelis Drugs 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K9/00—Medicinal preparations characterised by special physical form
- A61K9/0012—Galenical forms characterised by the site of application
- A61K9/0019—Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K9/00—Medicinal preparations characterised by special physical form
- A61K9/10—Dispersions; Emulsions
- A61K9/127—Liposomes
- A61K9/1271—Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K9/00—Medicinal preparations characterised by special physical form
- A61K9/14—Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
- A61K9/19—Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles lyophilised, i.e. freeze-dried, solutions or dispersions
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K9/00—Medicinal preparations characterised by special physical form
- A61K9/48—Preparations in capsules, e.g. of gelatin, of chocolate
- A61K9/50—Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
- A61K9/51—Nanocapsules; Nanoparticles
- A61K9/5107—Excipients; Inactive ingredients
- A61K9/5123—Organic compounds, e.g. fats, sugars
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/06—Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
- A61K47/28—Steroids, e.g. cholesterol, bile acids or glycyrrhetinic acid
Definitions
- vaccines containing antigens are typically not immunogenic enough to generate rapid and prolonged immunity. This can sometimes be overcome with the use of an adjuvant to boost the immune response towards an antigen.
- adjuvants There are generally two broad categories of adjuvants: delivery systems and immune-stimulants.
- the delivery system of a vaccine can act as an adjuvant by providing stability and prolonged interaction of the antigen with the immune system.
- Vaccine compositions may also incorporate molecular compounds with immune- stimulatory activity as adjuvants with the aim of further enhancing immunogenicity of the vaccine by directly activating cells of the immune system.
- Immune-stimulant adjuvants are defined molecular agonists that are recognized by the immune system via specialized receptors, for example polyI:C polynucleotide and lipid- based adjuvants such as PAM 3 CSK4 stimulate distinct Toll-like receptors. Immune-stimulants can activate the immune system and also direct the type of immune response generated towards a vaccine antigen.
- the effectiveness of many vaccines is correlated to the generation of antibodies; however, for other types of vaccines a strong cytotoxic immune response primarily mediated by CD8+ T cells is desired.
- the type of immune response generated towards a vaccine antigen can be manipulated by including immune-stimulants that activate particular receptors found on immune cells that can initiate these responses through generation of cytokines and chemokines.
- Differences in the properties of immune-stimulants such as polyI:C polynucleotide and lipid-based adjuvants can complicate their incorporation into pharmaceutical compositions.
- a method for preparing a pharmaceutical composition comprising: (a) providing: (i) a first dried preparation comprising at least one therapeutic agent, a lipid, and a polyI:C polynucleotide, and (ii) a second dried preparation comprising a lipid-based adjuvant and a lipid; (b) combining the first dried preparation with a hydrophobic carrier to produce a reconstituted preparation; and (c) combining the reconstituted preparation with the second dried preparation to produce a pharmaceutical composition.
- a method for preparing a pharmaceutical composition comprising: (a) providing: (i) a first dried preparation comprising at least one therapeutic agent, a lipid, and a polyI:C polynucleotide, and (ii) a second dried preparation comprising a lipid-based adjuvant and a lipid; (b) combining the second dried preparation with a hydrophobic carrier to produce a reconstituted preparation; and (c) combining the reconstituted preparation with the first dried preparation to produce a pharmaceutical composition.
- a pharmaceutical composition prepared by the method as described herein.
- a method of inducing an antibody and/or CTL immune response in a subject comprising administering to the subject the pharmaceutical composition as described herein.
- a use of the pharmaceutical composition as described herein for inducing an antibody and/or CTL immune response in a subject there is provided a method of delivering a therapeutic agent to a subject comprising administering to the subject the pharmaceutical composition as described herein.
- a use of the pharmaceutical composition as described herein for delivering a therapeutic agent to a subject comprising administering to the subject the pharmaceutical composition as described herein.
- kits for preparing the pharmaceutical composition as described herein comprising: a container comprising a first dried preparation prepared by the method as described herein; a container comprising second dried preparation prepared by the method as described herein; and a container comprising a hydrophobic carrier.
- Poly I:C polynucleotide is an oligonucleotide molecule, composed of either DNA or RNA nucleosides, which interacts with Toll-like receptors (TLRs) that can detect nucleic acids, such as TLR3.
- TLRs Toll-like receptors
- Lipid-based adjuvants such as PAM 2 Cys and PAM 3 Cys interact with TLR1/2. Stimulating multiple TLR receptors at once has been reported to have an additive or synergistic activating effect on dendritic cells.
- poly I:C polynucleotide and a lipid-based adjuvant such as PAM2Cys or PAM3Cys in a pharmaceutical composition that can ensure they engage with TLRs on the same cell can provide a potent activating effect to the immune system.
- Optimal doses of each adjuvant may need to be confirmed for different applications. For example, vaccines for cancer therapy may need stronger immune stimulation than vaccines for prevention of infectious disease.
- the lipopeptides PAM 2 Cys and PAM 3 Cys comprise a lipid palmitic acid moiety conjugated to a cysteine (Cys) residue that may be extended into a peptide with polar residues such as serine (Ser) and/or lysine (Lys) with a positive surface charge.
- Cys cysteine
- polyI:C polynucleotides are made up of single or double stranded molecules containing inosinic acid residues (I) and cytidylic acid residues (C) with a negative surface charge. If needed, these two distinct immune stimulators can be complexed together by electrostatic interactions to a certain extent with concentration restrictions.
- any one of these immune stimulators may lead to a pH shift and cause unwanted precipitation during formulation and/or in the finished product, constraining the ability to provide desired concentrations of lipid- based adjuvant and polyI:C polynucleotide in a pharmaceutical composition.
- Using current methods it is only possible to combine polyI:C polynucleotides and PAM 2 Cys and PAM 3 Cys lipid-based adjuvants in a single water-free formulation up to about 0.5 mg/mL concentration.
- Embodiments of the present invention provide methods of fully incorporating both polyI:C polynucleotide and lipid-based adjuvant (such as PAM 2 Cys and PAM 3 Cys) along with therapeutic agents that overcomes concentration limitations, irrespective of the nature of therapeutic agents and formulation buffer/pH used to prepare the pharmaceutical composition.
- polyI:C polynucleotide and lipid-based adjuvant such as PAM 2 Cys and PAM 3 Cys
- PolyI:C polynucleotide are polynucleotide molecules (either RNA or DNA or a combination of DNA and RNA) containing inosinic acid residues (I) and cytidylic acid residues (C), and which induce the production of inflammatory cytokines, such as interferon.
- the polyI:C polynucleotide is double-stranded. In double-stranded embodiments, they are typically composed of one strand consisting entirely of cytosine-containing nucleotides and one strand consisting entirely of inosine-containing nucleotides, although other configurations are possible.
- each strand may contain both cytosine-containing and inosine-containing nucleotides.
- the polyI:C polynucleotide is single- stranded. In single-stranded embodiments, the strand comprises both cytosine-containing and inosine-containing nucleotides. In some instances, either or both strand may additionally contain one or more non-cytosine or non-inosine nucleotides.
- the polyI:C polynucleotide may be a single-stranded molecule containing inosinic acid residues (I) and cytidylic acid residues (C).
- the single-stranded polyI:C polynucleotide may be a sequence of repeating dIdC.
- the sequence of the single-stranded polyI:C may be a 26-mer sequence of (IC) 13 , i.e. ICICICICICICICICICICICICIC.
- IC ICICICICICICICICICICICICIC
- a “polyI:C” or “polyI:C polynucleotide” is a double- or single-stranded polynucleotide molecule (RNA or DNA or a combination of DNA and RNA), each strand of which contains at least 6 contiguous inosinic or cytidylic acid residues, or 6 contiguous residues selected from inosinic acid and cytidylic acid in any order (e.g. IICIIC or ICICIC).
- PolyI:C polynucleotides will typically have a length of about 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 25, 26, 28, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 150, 200, 250, 300, 500, 1000 or more nucleotides.
- Each strand of a double-stranded polyI:C polynucleotide may be a homopolymer of inosinic or cytidylic acid residues, or each strand may be a heteropolymer containing both inosinic and cytidylic acid residues.
- the polymer may be interrupted by one or more non-inosinic or non-cytidylic acid residues (e.g. uridine), provided there is at least one contiguous region of 6 I, 6 C or 6 I/C residues as described above.
- each strand of a polyI:C polynucleotide will contain no more than 1 non-I/C residue per 6 I/C residues, more preferably, no more than 1 non-I/C residue per every 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 or 30 I/C residues.
- the inosinic acid or cytidylic acid (or other) residues in the polyI:C polynucleotide may be derivatized or modified as is known in the art, provided the ability of the polyI:C polynucleotide to promote the production of an inflammatory cytokine, such as interferon, is retained.
- Non-limiting examples of derivatives or modifications include e.g. azido modifications, fluoro modifications, or the use of thioester (or similar) linkages instead of natural phosphodiester linkages to enhance stability in vivo.
- the polyI:C polynucleotide may also be modified to e.g. enhance its resistance to degradation in vivo by e.g.
- lipid-based adjuvant is an adjuvant that comprises at least one lipid moiety or lipid component.
- lipid moiety or “lipid component” refers to any fatty acid (e.g. fatty acyls) or derivative thereof, including for example triglycerides, diglycerides, and monoglycerides.
- Exemplary fatty acids include, without limitation, palmitoyl, myristoyl, stearoyl and decanoyl groups or any C2 to C30 saturated or unsaturated fatty acyl group, preferably any C14 to C22 saturated or unsaturated fatty acyl group, and more preferably a C16 saturated or unsaturated fatty acyl group.
- lipid-based adjuvant encompasses adjuvants comprising a fatty acyl group or derivative thereof.
- Lipid-based adjuvants contain at a minimum at least one lipid moiety, or a synthetic/semi-synthetic lipid moiety analogue, which can be coupled onto an amino acid, an oligopeptide or other molecules (e.g. a carbohydrate, a glycan, a polysaccharide, biotin, Rhodamine, etc.).
- the lipid-based adjuvant may be, for example, a lipoamino acid, a lipopeptide, a lipoglycan, a lipopolysaccharide or a lipoteichoic acid.
- a lipid moiety or a structure containing a lipid moiety can be coupled covalently or non-covalently to an antigen to create antigenic compounds with built-in adjuvanting properties.
- the lipid-based moiety may comprise a cation (e.g. nickel) to provide a positive charge for non-covalent coupling.
- the lipid moiety or lipid component may be naturally occurring, such as for example a cell-wall component (e.g. lipoprotein) from a Gram-positive or Gram-negative bacteria, Rhodopseudomonas viridis, or mycoplasma.
- the lipid moiety or lipid component may be synthetic or semi-synthetic
- the lipid-based adjuvant may comprise palmitic acid (PAM) as at least one of the lipid moieties or components of the adjuvant.
- PAM palmitic acid
- Such lipid-based adjuvants are referred to herein as a “palmitic acid adjuvant”. Palmitic acid is a low molecular weight lipid found in the immunologically reactive Braun’s lipoprotein of Escherichia coli. Other common chemical names for palmitic acid include, for example, hexadecanoic acid in IUPAC nomenclature and 1-Pentadecanecarboxylic acid.
- a palmitic acid adjuvant contains at a minimum at least one palmitic acid moiety, which can be coupled onto an amino acid, an oligopeptide or other molecules.
- a palmitic acid moiety or a structure containing palmitic acid can be coupled covalently or non-covalently to an antigen to create antigenic compounds with built-in adjuvanting properties.
- the palmitic acid moiety or a chemical structure containing palmitic acid can be conjugated to a cysteine peptide (Cys) to allow for various structural configurations of the adjuvant, including linear and branched structures.
- the cysteine residue has been commonly extended by polar residues such as Serine (Ser) and/or lysine (Lys) at the C terminus to create adjuvant compounds with improved solubility.
- Palmitic acid containing adjuvant compounds could be admixed with an antigen, associated with antigen through non-covalent interactions, or alternatively covalently linked to an antigen, either directly or with the use of a linker/spacer, to generate enhanced immune responses.
- an antigen associated with antigen through non-covalent interactions
- an antigen either directly or with the use of a linker/spacer
- two palmitic acid moieties are attached to a glyceryl backbone and a cysteine residue to create dipalmitoyl-S-glyceryl-cysteine (PAM2Cys) or tripalmitoyl-S-glyceryl-cysteine (PAM3Cys), which can also be used in multiple configurations as described above.
- PAM2Cys dipalmitoyl-S-glyceryl-cysteine
- PAM3Cys tripalmitoyl-S-glyceryl-cysteine
- the lipid-based adjuvant is any type of adjuvant comprising a palmitic acid moiety or component.
- lipid-based adjuvant is a lipopeptide comprising one or more palmitic acid moieties.
- the palmitic acid moiety may be modified or manipulated to improve its stability in vitro or in vivo, enhance its binding to receptors (such as for example toll-like receptors as described below) or enhance its biological activity.
- the palmitic acid adjuvant comprises PAM2Cys.
- the palmitic acid adjuvant comprises PAM 3 Cys.
- the palmitic acid adjuvant comprises PAM 2 -Cys-Ser-(Lys)4 or PAM3-Cys-Ser-(Lys)4.
- the palmitic acid adjuvant is an analog of PAM2-Cys-Ser- (Lys)4 or PAM3-Cys-Ser-(Lys)4 including, without limitation, PAM3Cys-SKKKK ( ⁇ - irradiated), R-PAM3Cys-SKKKK S-PAM3Cys-SKKKK, PAM3Cys-SKKKK(Biotin-Aca-Aca), PAM 3 Cys-SKKKK(Fluorescein-Aca-Aca), PAM 3 Cys-SKKKK, PAM 3 Cys-SKKKK-FLAG-tag, PAM3Cys-SSNAKIDQLSSDVQT, PAM3Cys-SSNKSTT
- the palmitic acid adjuvant or analog thereof may be used as stereochemically defined compounds or as a mixture of stereoisomers.
- the lipid-based adjuvant is PAM 3 -Cys-Ser-(Lys)4: that act as TLR agonists may also be used as the lipid-based adjuvant disclosed herein, including without limitation the palmitic acid adjuvants and analogs described above and synthetic diacylated lipoprotein FSL-1 available from InvivoGen (San Diego, California, USA) and EMC Microcollections GmbH (Germany).
- FSL-1 (Pam2CGDPKHPKSF) is a synthetic lipoprotein that represents the N-terminal part of the 44-kDa lipoprotein LP44 of Mycoplasma salivarium.
- FSL-1 comprises PAM 2 Cys and has a similar framework structure as macrophage activating lipopeptide-2 (MALP-2), a Mycoplasma fermentans derived lipopeptide.
- MALP-2 macrophage activating lipopeptide-2
- the lipid-based adjuvant comprises FSL-1 or MALP-2, or the lipid-based adjuvant is FSL-1 or MALP-2.
- FSL-1 or MALP-2 may be used as stereochemically defined compounds or as a mixture of stereoisomers.
- the FSL-1 or MALP- 2 may be labelled (e.g.
- lipid-based adjuvants that comprise palmitic acid may include substructures of TLR2 ligands such as monoacylated lipopeptides. Without limitation, these may include, for example, Pam-Dhc-SKKKK, Pam-CSKKKK, Pam-Dhc-GDPKHPKSF or Pam-CGDPKHPKSF (EMC Microcollections).
- therapeutic agent is any molecule, substance or compound that is capable of providing a therapeutic activity, response or effect in the treatment or prevention of a disease, disorder or condition, including diagnostic and prophylactic agents.
- a “first therapeutic agent” is any one or more therapeutic agents which are used in the preparation of the non-sized lipid vesicle particle preparation (i.e. incorporated in the methods before the step of sizing the non-sized lipid vesicle preparation).
- a “second therapeutic agent” is any one or more therapeutic agents which are used in the methods herein after preparation of the sized lipid vesicle particle preparation (i.e.
- lipids may first be dissolved and mixed in an organic solvent. In embodiments where different types of lipid are used, this step will allow a homogenous mixture of the lipids to be formed. In an embodiment, these steps may be carried out in chloroform, chloroform:methanol mixtures, tertiary butanol or cyclohexane. In an embodiment, the lipids are prepared at 10-20mg lipid/mL organic solvent; however, higher or lower concentrations may also be used. In some embodiments, after mixing, the organic solvent is removed (e.g. by evaporation) to yield a lipid film.
- the lipid film may then be frozen and lyophilized to yield a dry lipid film.
- the dry lipid film may then be hydrated with an aqueous solution containing therapeutic agents to provide a non-sized lipid vesicle particle preparation.
- the step of hydration may be performed with shaking and/or mixing (e.g. at 300 RPM for about 1 hour).
- an aqueous solution of lipids may be combined with a solution containing one or more solubilized therapeutic agents.
- one or more dry therapeutic agents may be added to, and solubilized in, the aqueous solution of lipids or sized lipid vesicle preparation. These embodiments may be performed with shaking and/or mixing (e.g.
- therapeutic agents are either solubilized in a solvent (e.g. aqueous or organic) prior to mixing with lipid vesicle particles or therapeutic agents are solubilized upon being mixed with the lipid vesicle particles.
- a solvent e.g. aqueous or organic
- therapeutic agents may be added as a dry powder to a solution containing lipid vesicle particles or both the lipid vesicle particles and dry therapeutic agents may be mixed together in a fresh solvent.
- the therapeutic agent is an antigen, a small molecule drug, an antibody or a functional fragment thereof, an antibody mimetic or a functional fragment thereof, an immunomodulatory agent, a polynucleotide encoding a polypeptide, or an interfering polynucleotide.
- the therapeutic agent is an antigen.
- the term “antigen” may be used interchangeably with “immunogen” and may refer to a pathogen, a part of a pathogen, or a molecule that is able to induce an adaptive antibody immune response and/or an adaptive cellular immune response that is specific to said antigen or a portion thereof.
- the term “antigen” may refer to a pathogen, a part of a pathogen, or a molecule that is specifically recognized by antibodies and/or immunoglobulin receptors of the adaptive antibody immune response and/or the adaptive cellular immune response.
- the term “peptide antigen” is an antigen as defined above that is a protein or a polypeptide.
- Adaptive antibody responses are mediated by B cells, which recognize antigen by specific binding of IgM immunoglobulins in their B cell receptors to the antigen. Activated B cells may mature into plasma cells and secrete soluble IgM, IgG, IgA, or IgE antibodies that specifically to bind to the antigen.
- T cell receptors specifically the immunoglobulin superfamily TCR-alpha/TCR-beta or TCR-gamma/TCR-delta heterodimers
- T cell receptors specifically the immunoglobulin superfamily TCR-alpha/TCR-beta or TCR-gamma/TCR-delta heterodimers
- Antibody and cellular adaptive immune responses are coordinated by CD4+ T helper cells, which recognize polypeptide antigen by specific binding of their T cell receptors (specifically the immunoglobulin superfamily TCR-alpha/TCR-beta or TCR-gamma/TCR-delta heterodimers) to peptides from the antigen that are displayed on MHC class I/II molecules on the surface of APC.
- T cell receptors specifically the immunoglobulin superfamily TCR-alpha/TCR-beta or TCR-gamma/TCR-delta heterodimers
- the immunoglobulin superfamily proteins in B cell receptors and antibodies (IgM, IgD, IgG, IgA, IgE), and in T cell receptors (TCR-alpha/TCR-beta, TCR-gamma/TCR-delta) that specifically bind to antigen are created randomly by processes of genetic recombination during B cell and T cell development.
- Each B cell and T cell expresses only one species of randomized immunoglobulin superfamily receptor with a specific binding recognition, and B cells and T cells that bind an antigen are clonally selected for expansion in the body during an immune response.
- the immunoglobulins in B cell receptors and antibodies may further diversify by somatic mutation and clonal selection for variants with high antigen binding.
- antibodies, B cell receptors, and T cell receptors are immunoglobulin superfamily molecules that enable the immune system to adapt to an antigen, thus forming an adaptive immune response.
- This adaptive immune response contrasts with innate immune responses that are mediated by conserved immune receptors, such as TLR or other pattern recognition receptors, that are unchanging and have fixed specificity for binding to specific pathogen- associated molecular patterns.
- the term “antigen” or “peptide antigen” may refer to a molecule that is specifically bound by antibodies, B cell receptors, and/or T cell receptors of the adaptive immune system during an active adaptive immune response.
- an antigen that is bound by an antibody, a B cell receptor, and/or a T cell receptor is called an “epitope”.
- Antibodies and B cell receptors may bind to an epitope on various types of molecules including polypeptides, polysaccharides, glycoproteins, and lipoproteins.
- T cell receptors bind to peptides that are derived from antigen, wherein the peptides are displayed on MHC class I/II molecules.
- the term “antigen” may refer to a molecule that comprises a B cell epitope and/or a T cell epitope.
- the antigen may include a polypeptide, a polysaccharide, a glycoprotein, a lipoprotein, a microorganism or a part thereof, such as a live, attenuated, inactivated or killed bacterium, virus or protozoan, or part thereof, an allergen, or an antigen derived from a cancer cell (such as a conserved cancer antigen or a neoantigen).
- the term “derived from” encompasses, without limitation: an antigen that is isolated or obtained directly from an originating source; a synthetic or recombinantly generated antigen that is identical or substantially related to an antigen from an originating source; or an antigen which is made from an antigen of an originating source or a fragment thereof.
- an antigen is “from” a source
- the term “from” may be equated with “derived from”.
- substantially related means that the antigen may have been modified by chemical, physical or other means (e.g. sequence modification), but that the resultant product remains capable of generating an immune response to the original antigen or to the disease or disorder associated with the original antigen.
- the term “antigen” also includes a polynucleotide that encodes a polypeptide that functions as an antigen.
- Nucleic acid-based vaccination strategies are known, wherein a vaccine composition that contains a polynucleotide is administered to a subject.
- the antigenic polypeptide encoded by the polynucleotide is expressed in the subject, such that the antigenic polypeptide is ultimately present in the subject.
- Viruses, or parts thereof, from which a peptide antigen may be derived include for example, and without limitation, Cowpoxvirus, Vaccinia virus, Pseudocowpox virus, herpes virus, Human herpesvirus 1, Human herpesvirus 2, Cytomegalovirus, Human adenovirus A-F, Polyomavirus, human papillomavirus (HPV), Parvovirus, Hepatitis A virus, Hepatitis B virus, Hepatitis C virus, human immunodeficiency virus (HIV), Seneca Valley virus (SVV), Orthoreovirus, Rotavirus, Ebola virus, parainfluenza virus, influenza virus (e.g.
- influenza virus H5N1 influenza virus, influenza A virus, influenza B virus, influenza C virus), Measles virus, Mumps virus, Rubella virus, Pneumovirus, respiratory syncytial virus, respiratory syncytial virus (RSV), Rabies virus, California encephalitis virus, Japanese encephalitis virus, Hantaan virus, Lymphocytic choriomeningitis virus, Coronavirus (e.g. Sars-Cov-2), Enterovirus, Rhinovirus, Poliovirus, Norovirus, Flavivirus, Dengue virus, West Nile virus, Yellow fever virus and varicella.
- the peptide antigen is derived from HPV.
- the HPV peptide antigen is one that is associated with HPV-related cervical cancer or HPV-related head and neck cancer.
- the peptide antigen is a peptide comprising the sequence RAHYNIVTF (HPV16E7 (H-2Db) peptide 49-57; R9F).
- the peptide antigen is a peptide comprising the sequence YMLNLGPET (HPV Y9T peptide).
- the peptide antigen is derived from HIV.
- the HIV peptide antigen may be derived from the V3 loop of HIV-1 gp120.
- the HIV peptide antigen may be RGP10 (RGPGRAFVTI).
- the peptide antigen may be AMQ9 (AMQMLKETI).
- AMQ9 peptide is the immunodominant MHC class I epitope of gag for mice of the H-2Kd haplotype.
- the peptide antigen is derived from RSV.
- the RSV virion a member of the genus Paramyxoviridae, is composed of a single strand of negative-sense RNA with 15,222 nucleotides.
- the nucleotides encode three transmembrane surface proteins (F, G and small hydrophobic protein or SH), two matrix proteins (M and M2), three nucleocapsid proteins (N, P and L), and two non-structural proteins (NS1 and NS2).
- the peptide antigen may be derived from any one or more of the RSV proteins.
- the peptide antigen may be derived from the SH protein of RSV or any other paramyxovirus, or a fragment thereof.
- the RSV peptide antigen may be any one or more of the RSV peptides described or disclosed in WO 2012/065997, incorporated herein by reference.
- Bacteria, or parts thereof, from which a peptide antigen may be derived include for example, and without limitation, Anthrax (Bacillus anthracis), Brucella, Bordetella pertussis, Candida, Chlamydia pneumoniae, Chlamydia psittaci, Cholera, Clostridium botulinum, Coccidioides immitis, Cryptococcus, Diphtheria, Escherichia coli O157: H7, Enterohemorrhagic Escherichia coli, Enterotoxigenic Escherichia coli, Haemophilus influenzae, Helicobacter pylori, Legionella, Leptospira, Listeria, Meningococcus, Mycoplasma pneumoniae, Mycobacterium, Pertussis, Pneumonia, Salmonella, Shigella, Staphylococcus, Streptococcus pneumoniae and Yersinia enterocolitica.
- Anthrax Bacteria,
- the peptide antigen is derived from a Bacillus anthracis.
- the peptide antigen may for example be derived from anthrax recombinant protective antigen (rPA) (List Biological Laboratories, Inc.; Campbell, CA) or anthrax mutant recombinant protective antigen (mrPA).
- rPA has an approximate molecular weight of 83,000 daltons (Da) and corresponds a cell binding component of the three-protein exotoxin produced by Bacillus anthracis.
- the protective antigen mediates the entry of anthrax lethal factor and edema factor into the target cell.
- the antigen may be derived from the sequence found under GenBank Accession number P13423, or any suitable sequence variant thereof.
- Protozoa, or parts thereof, from which a peptide antigen may be derived include for example, and without limitation, the genus Plasmodium (Plasmodium falciparum, Plasmodium malariae, Plasmodium vivax, Plasmodium ovale or Plasmodium knowlesi), which causes malaria.
- the peptide antigen is derived from a Plasmodium species.
- the peptide antigen may be derived from the circumsporozoite protein (CSP), which is a secreted protein of the sporozoite stage of the malaria parasite (Plasmodium sp.).
- CSP circumsporozoite protein
- the amino-acid sequence of CSP consists of an immunodominant central repeat region flanked by conserved motifs at the N- and C-termini that are implicated in protein processing as the parasite travels from the mosquito to the mammalian vector.
- the structure and function of CSP is highly conserved across the various strains of malaria that infect humans, non-human primates and rodents.
- the peptide antigen derived from CSP is a malaria virus-like particle (VLP) antigen which comprises circumsporozoite T and B cell epitopes displayed on the woodchuck hepatitis virus core antigen.
- VLP malaria virus-like particle
- the peptide antigen may be derived from a cancer or tumor-associated protein, such as for example, a membrane surface-bound cancer antigen.
- the cancer may be one that is caused by a pathogen, such as a virus.
- Viruses linked to the development of cancer include, but are not limited to, human papillomaviruses (HPV), John Cunningham virus (JCV), Human herpes virus 8, Epstein Barr Virus (EBV), Merkel cell polyomavirus, Hepatitis C Virus and Human T cell leukaemia virus-1.
- HPV human papillomaviruses
- JCV John Cunningham virus
- EBV Epstein Barr Virus
- Merkel cell polyomavirus Hepatitis C Virus
- Hepatitis C Virus Human T cell leukaemia virus-1.
- the peptide antigen may be derived from a virus that is linked to the development of cancer.
- the peptide antigen is a cancer-associated antigen.
- Many cancer or tumor-associated proteins are known in the art such as for example, and without limitation, those described in WO 2016/176761, incorporated by reference herein.
- the peptide antigen is one or more survivin antigens.
- Survivin also called baculoviral inhibitor of apoptosis repeat-containing 5 (BIRC5), is a protein involved in the negative regulation of apoptosis.
- the peptide antigen is any peptide, polypeptide or variant thereof derived from a survivin protein, or a fragment thereof.
- the survivin peptide antigen may comprise the full length survivin polypeptide.
- the survivin peptide antigen may be a survivin peptide comprising a fragment of any length of the survivin protein.
- Exemplary embodiments include a survivin peptide that comprises at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 amino acid residues.
- the survivin peptide consists of a heptapeptide, an octapeptide, a nonapeptide, a decapeptide or an undecapeptide, consisting of 7, 8, 9, 10, 11 consecutive amino acid residues of the survivin protein, respectively.
- Particular embodiments of the survivin antigen include survivin peptides of about 9 or 10 amino acids.
- Survivin peptide antigens also encompass variants and functional equivalents of natural survivin peptides.
- Variants or functional equivalents of a survivin peptide encompass peptides that exhibit amino acid sequences with differences as compared to the specific sequence of the survivin protein, such as one or more amino acid substitutions, deletions or additions, or any combination thereof. The difference may be measured as a reduction in identity as between the survivin protein sequence and the survivin peptide variant or survivin peptide functional equivalent.
- a pharmaceutical composition of the present invention may include any one or more of the survivin peptides, survivin peptide variants or survivin peptide functional equivalents disclosed in WO 2004/067023; WO 2006/081826 or WO 2016/176761, each of which is incorporated by reference herein.
- the survivin peptide antigen may be any one or more of FEELTLGEF, FTELTLGEF, LTLGEFLKL, LMLGEFLKL, RISTFKNWPF, RISTFKNWPK, STFKNWPFL, and/or LPPAWQPFL.
- the peptide antigen is a neoantigen.
- the term “neoantigen” refers to a class of tumor antigens which arise from tumor-specific mutations in an expressed protein. The neoantigen can be derived from any cancer, tumor or cell thereof.
- neoantigens the term “derived from” as used herein encompasses, without limitation: a neoantigen that is isolated or obtained directly from an originating source (e.g. a subject); a synthetic or recombinantly generated neoantigen that is identical in sequence to a neoantigen from an originating source; or a neoantigen which is made from a neoantigen of an originating source or a fragment thereof.
- the mutations in the expressed protein that create the neoantigen may be patient-specific. By “patient-specific”, it is meant that the mutation(s) are unique to an individual subject. However, it is possible that more than one subject will share the same mutation(s).
- neoantigen may comprise one or more neoepitopes.
- epitope refers to a peptide sequence which can be recognized by the immune system, specifically by antibodies, B cells or T cells.
- a “neoepitope” is an epitope of a neoantigen which comprises a tumor-specific mutation as compared to the native amino acid sequence.
- neoepitopes may be identified by screening neoantigens for anchor residues that have the potential to bind patient HLA.
- the neoepitopes are normally ranked using algorithms, such as NetMHC, that can predict peptide binding to HLA.
- a "T-cell neoepitope” is to be understood as meaning a mutated peptide sequence which can be bound by the MHC molecules of class I or II in the form of a peptide-presenting MHC molecule or MHC complex.
- the T-cell neoepitope should typically be one that is amenable to recognition by T cell receptors so that a cell-mediated immune response can occur.
- a "B-cell neoepitope” is to be understood as meaning a mutated peptide sequence which can be recognized by B cells and/or by antibodies.
- At least one of the neoepitopes of the neoantigen is a patient-specific neoepitope.
- patient-specific neoepitope it is meant that the mutation(s) in the neoepitope are unique to an individual subject. However, it is possible that more than one subject will share the same mutation(s). Thus, a “patient-specific neoepitope” may be shared by a small or large sub-population of subjects.
- the neoantigen may be derived from a mutated gene or protein that has previously been associated with cancer phenotypes, such as for example tumor suppressor genes (e.g.
- the neoantigen may comprise or consist of the neoantigens disclosed in Castle et al. (2012) Exploiting the Mutanome for Tumor Vaccination. Cancer Res, 72(5): 1081-1091, incorporated by reference herein.
- the neoantigen may be one or more of the Mut1-50 neoantigens disclosed in Table 1 of Castle 2012, or a neoantigen of the same or related protein (e.g. a human homologue).
- the neoantigen may be one or more of Mut25 (STANYNTSHLNNDVWQIFENPVDWKEK), Mut30 (PSKPSFQEFVDWENVSPELNSTDQPFL) and Mut44 (EFKHIKAFDRTFANNPGPMVVFATPGM), or a neoantigen of the same or related protein (e.g. a human homologue).
- Mut25 STANYNTSHLNNDVWQIFENPVDWKEK
- Mut30 PSKPSFQEFVDWENVSPELNSTDQPFL
- Mut44 EFKHIKAFDRTFANNPGPMVVFATPGM
- a neoantigen of the same or related protein e.g. a human homologue
- the antigen may be a peptide derived from the ectodomain of the small hydrophobic protein of respiratory syncytial virus (RSV) as disclosed in WO2012/065997, incorporated herein by reference.
- RSV respiratory syncytial virus
- the RSV antigen may be a peptide with the sequence NKLCEYNVFHNKTFELPRARVNT, NKLSEHKTFCNNTLELGQMHQINT, or NKLCDFNDHHTNSLDIRTRLRNDTQLITRAHEGSINQSSN, or a portion or variant thereof
- the antigen may be a peptide derived from the survivin protein as disclosed in WO2004/067023 and WO2006/081826, incorporated herein by reference.
- the survivin antigen may be one or more peptides of the sequence FEELTLGEF, FTELTLGEF, LTLGEFLKL, LMLGEFLKL, RISTFKNWPF, RISTFKNWPK, STFKNWPFL, or LPPAWQPFL, or a portion or a variant thereof.
- the antigen may be a fusion peptide (FP) antigen comprising the sequence RAHYNIVTF (HPV16E7 (H-2Db) peptide 49-57) fused to the universal T-helper epitope is PADRE (pan-DR epitope) comprising the peptide sequence AKXVAAWTLKAAA, wherein X may be phenylalanine or cyclohexylalanine.
- the therapeutic agent is one or more peptide antigens as described herein.
- the peptide antigen is a synthetically produced polypeptide.
- the peptide antigen may be a polypeptide of any length.
- the peptide antigen may be 5 to 120 amino acids in length, 5 to 100 amino acids in length, 5 to 75 amino acids in length, 5 to 50 amino acids in length, or 5 to 30 amino acids in length. In an embodiment, the peptide antigen may be 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49 or 50 amino acids in length. In an embodiment, the peptide antigen is 20 to 30 amino acids in length. In an embodiment, the peptide antigen is 27 amino acids in length. In an embodiment, the peptide antigen is 8 to 40 amino acids in length.
- the peptide antigen is 9 or 10 amino acids in length.
- the therapeutic agent is a small molecule drug.
- small molecule drug refers an organic or inorganic compound that may be used to treat, cure, prevent or diagnose a disease, disorder or condition.
- small molecule refers to a low molecular weight compound which may be synthetically produced or obtained from natural sources and has a molecular weight of less than 2000 Daltons (Da), less than 1500 Da, less than 1000 Da, less than 900 Da, less than 800 Da, less than 700 Da, less than 600 Da or less than 500 Da.
- a small molecule drug is typically a chemically manufactured active substance or compound (i.e.
- small molecule drug does not encompass larger structures, such as polynucleotides, proteins, and polysaccharides, which are made by a biological process.
- small molecule may refer to compounds or molecules that selectively bind specific biological macromolecules and act as an effector, altering the activity or function of the target.
- a small molecule drug is a substance or compound that regulates a biological process in the body of a subject, and more particularly within a cell.
- a small molecule drug may exert its activity in the form in which it is administered, or the small molecule drug may be a prodrug.
- small molecule drug encompasses both the active form and the prodrug.
- prodrug refers to a compound or substance that, under physiological conditions, is converted into the therapeutically active agent.
- a prodrug is a compound or substance that, after administration, is metabolized in the body of a subject into the pharmaceutically active form (e.g. by enzymatic activity in the body of the subject).
- a common method for making a prodrug is to include selected moieties that are hydrolyzed under physiological conditions to reveal the pharmaceutically active form.
- Many small molecule drugs are known in the art and are used as active ingredients in medicaments. The skilled person is aware of numerous small molecule drugs such as those disclosed in the online DrugBank database.
- a small molecule drug may include a cytotoxic agent, an anti-cancer agent, an anti- tumor agent, a chemotherapeutic agent, an anti-neoplastic agent, an antiviral agent, an antibacterial agent, an anti-inflammatory agent, an immunomodulatory agent (e.g. a cytokine or a chemokine), an immune response checkpoint agent, a biological response modifier, a prodrug, a ligand, an analgesic, a radiopharmaceutical, a radioisotope or a dye for visual detection.
- a cytotoxic agent may be an agent that kills a target cell by necrosis or apoptosis.
- a cytotoxic agent may include epacadostat, cyclophosphamide, rapamycin, ifosfamide, afosfamide, melphalan, bendamustine, uramustine, palifosfamide, chlorambucil, busulfan, 4-hydroxycyclophosphamide, bis-chloroethylnitrosourea (BCNU), mitomycin C, yondelis, procarbazine, dacarbazine, temozolomide, cisplatin, carboplatin, oxaliplatin, acyclovir, gemcitabine, 5-fluorouracil, cytosine arabinoside, ganciclovir, camptothecin, topotecan, irinotecan, doxorubicin, daunorubicin, epirubicin, idarubicin, etoposide, teniposide, mitoxantrone, valproic acid, vorin
- a small molecule drug may include an checkpoint agent.
- a “checkpoint agent” refers to any compound or molecule that totally or partially modulates (e.g. activates or inhibits) the activity or function of one or more checkpoint molecules (e.g. proteins). Checkpoint molecules regulate various cellular processes.
- a small molecule drug may include a checkpoint agent that is an inhibitor of Programmed Death-Ligand 1 (PD-L1, also known as B7-H1, CD274), Programmed Death 1 (PD-1, CD279), CTLA-4 (CD154), PD-L2 (B7-DC, CD273), LAG3 (CD223), TIM3 (HAVCR2, CD366), 41BB (CD137), 2B4, A2aR, B7H1, B7H3, B7H4, B- and T-lymphocyte attenuator (BTLA), CD2, CD27, CD28, CD30, CD33, CD40, CD70, CD80, CD86, CD160, CD226, CD276, DR3, GAL9, GITR, HVEM, IDO1, IDO2, ICOS (inducible T cell costimulator), Killer inhibitory receptor (KIR), LAG-3, LAIR1, LIGHT, MARCO (macrophage receptor with collageneous structure), phosphatidylserine (PS
- a small molecule drug may include a cell-penetrating peptide, a peptide transduction domain, or a dendritic cell peptide, used as molecular shuttles that can transport other molecules or ions from one location to another.
- the therapeutic agent is an antibody, a functional equivalent of an antibody or a functional fragment of an antibody.
- an “antibody” refers to a polypeptide or protein that consists of or comprises antibody domains, which are understood as constant and/or variable domains of the heavy and/or light chains of immunoglobulins, with or without a linker sequence. Antibody domains may be of native structure or modified by mutagenesis or derivatization, e.g.
- An antibody may comprise a complete (i.e. full-length) immunoglobulin molecule, including e.g. polyclonal, monoclonal, chimeric, humanized and/or human versions having full length heavy and/or light chains.
- the term “antibody” encompasses any and all isotypes and subclasses, including without limitation the major classes of IgA, IgD, IgE, IgG and IgM, and the subclasses IgG1, IgG2, IgG3, IgG4, IgA1 and IgA2.
- An antibody may be one that is naturally occurring or one that is prepared by any means available to the skilled person, such as for example by using immunoglobulin gene fragment recombinatorial processes.
- An antibody may be a monoclonal antibody, a polyclonal antibody, a chimeric antibody, a humanized antibody, a human antibody or a fully human antibody.
- a “chimeric antibody” as used herein refers to a recombinant protein that contains the variable domains (including the complementarity determining regions (CDRs)) of an antibody derived from one species, such for example a rodent, while the constant domains of the antibody are derived from a different species, such as a human.
- CDRs complementarity determining regions
- the constant domains of the chimeric antibody may be derived from that of an animal, such as for example a cat or dog.
- a “humanized antibody” as used herein refers to a recombinant protein in which the CDRs from an antibody from one species; e.g., a rodent, are transferred from the heavy and light variable chains of the rodent antibody into human heavy and light variable domains, including human framework region (FR) sequences.
- the constant domains of the humanized antibody are likewise derived from a human antibody.
- a “human antibody” as used herein refers to an antibody obtained from transgenic animals (e.g. mice) that have been genetically engineered to produce specific human antibodies in response to antigenic challenge.
- elements of the human heavy and light chain loci are introduced into strains of mice derived from embryonic stem cell lines that contain targeted disruptions of the endogenous heavy chain and light chain loci.
- the transgenic animal can synthesize human antibodies specific for human antigens, and the animal can be used to produce human antibody-secreting hybridomas.
- a fully human antibody also can be constructed by genetic or chromosomal transfection methods, as well as phage display technology, all of which are known in the art.
- the term “functional fragment”, with respect to an antibody refers to an antigen-binding portion of an antibody. In this context, by “functional” it is meant that the fragment maintains its ability to bind to the target antigen.
- Functional fragments of antibodies include a portion of an antibody such as a F(ab') 2 , a F(ab) 2 , a Fab', a Fab, a Fab2, a Fab3, and single domain antibody. Regardless of structure, a functional fragment of an antibody binds with the same antigen that is recognized by the intact antibody.
- the term “functional fragment”, in relation to antibodies, also includes isolated fragments consisting of the variable regions, such as the “Fv” fragments consisting of the variable regions of the heavy and light chains and recombinant single chain polypeptide molecules in which light and heavy chain variable regions are connected by a peptide linker (“scFv proteins”).
- Antibody fragments can be incorporated into single domain antibodies (e.g.
- nanobodies single-chain antibodies
- maxibodies evibodies
- minibodies intrabodies
- diabodies triabodies
- tetrabodies vNAR, bis-scFv and other like structures.
- Another form of a functional fragment is a peptide comprising one or more CDRs of an antibody or one or more portions of the CDRs, provided the resultant peptide retains the ability to bind the target antigen.
- Antibodies for therapeutic use are known in the art such as, for example, anti-CTLA4 antibodies (ipilimumab, tremelimumab, BN- 13, UC10-4F10-11, 9D9 or 9H10), anti-PD1 antibodies (pembrolizumab, nivolumab, pidilizumab, AMP-224, RMP1-4 or J43), and anti-PDL1 antibodies (tezolizumab, avelumab, BMS-936559 or durvalumab).
- the therapeutic agent is an antibody mimetic, a functional equivalent of an antibody mimetic, or a functional fragment of an antibody mimetic.
- antibody mimetic refers to compounds which, like antibodies, can specifically and/or selectively bind antigens or other targets, but which are not structurally related to antibodies.
- Antibody mimetics are usually artificial peptides or proteins and are typically smaller than antibodies, with a molar mass of about 3-20 kDa (whereas antibodies are generally about 150 kDa).
- Non-limiting examples of antibody mimetics include peptide aptamers, affimers, affilins, affibodies, affitins, alphabodies, anticalins, avimers, DARPins, fynomers, Kunitz domain peptides, nanoCLAMPs, monobodies, affinity reagents and scaffold proteins.
- the term “functional fragment”, with respect to an antibody mimetic, refers any portion or fragment of an antibody mimetic that maintains the ability to bind to its target molecule.
- the functional fragment of an antibody mimetic may be, for example, a portion of any of the antibody mimetics as described herein.
- a “functional equivalent” in the context of an antibody mimetic refers to a polypeptide or other compound or molecule having similar binding characteristics to an antibody mimetic, but not necessarily being a recognizable “fragment” of an antibody mimetic.
- the therapeutic agent is an immunomodulatory agent.
- an “immunomodulatory agent” is a molecule or compound that modulates the activity and/or effectiveness of an immune response.
- “Modulate”, as used herein, means to enhance (upregulate), suppress (downregulate), direct, redirect or reprogram an immune response.
- the term “modulate” is not intended to mean activate or induce. By this, it is meant that the immunomodulatory agent modulates (enhances, reduces or directs) an immune response that is activated, initiated or induced by a particular antigen, but the immunomodulatory agent is not itself the antigen against which the immune response is directed, nor is the immunomodulatory agent derived from that antigen.
- An immunomodulatory agent that enhances the immune response may be selected from cytokines (e.g.
- interleukins and interferons certain interleukins and interferons
- stem cell growth factors include lymphotoxins, co-stimulatory molecules, hematopoietic factors, colony stimulating factors, erythropoietins, thrombopoietins, and the like, and synthetic analogs of these molecules.
- An immunomodulatory agent may include: lymphotoxins, such as tumor necrosis factor (TNF); hematopoietic factors, such as interleukin (IL); colony stimulating factor, such as granulocyte-colony stimulating factor (G-CSF) or granulocyte macrophage-colony stimulating factor (GM-CSF); interferon, such as interferons-alpha, -beta or –lamda; and stem cell growth factor, such as that designated "SI factor".
- lymphotoxins such as tumor necrosis factor (TNF); hematopoietic factors, such as interleukin (IL); colony stimulating factor, such as granulocyte-colony stimulating factor (G-CSF) or granulocyte macrophage-colony stimulating factor (GM-CSF); interferon, such as interferons-alpha, -beta or –lamda; and stem cell growth factor, such as that designated "SI factor”.
- cytokines include hormones, such as human growth hormone, N-methionyl human growth hormone, and bovine growth hormone; parathyroid hormone; thyroxine; insulin; proinsulin; relaxin; prorelaxin; glycoprotein hormones, such as follicle stimulating hormone (FSH), thyroid stimulating hormone (TSH), and luteinizing hormone (LH); hepatic growth factor; prostaglandin, fibroblast growth factor; prolactin; placental lactogen, OB protein; tumor necrosis factor-alpha and -beta; mullerian-inhibiting substance; mouse gonadotropin-associated peptide; inhibin; activin; vascular endothelial growth factor; integrin; thrombopoietin (TPO); nerve growth factors, such as NGF-beta; platelet-growth factor; transforming growth factors (TGFs), such as TGF-alpha and TGFP; insulin-like growth factor-I and -II; erythro
- An immunomodulatory agent may be an immune costimulatory molecule agonist.
- Immune costimulatory molecules are signaling proteins that play a role in regulating immune response. Some immune costimulatory molecules are receptors located on the surface of a cell that respond to extracellular signaling. When activated, immune costimulatory molecules produce a pro-inflammatory response that can include suppression of regulatory T cells and activation of cytotoxic or killer T cells.
- Exemplary immune costimulatory molecules include CD27, CD28, CD40, CD122, CD137, CD137/4-1BB, ICOS, IL-10, OX40 TGF-beta, TOR receptor, and glucocorticoid-induced TNFR-related protein GITR.
- An immune costimulatory molecule agonist may therefore be any compound, molecule or substance that is an agonist of a costimulatory immune molecule as described herein.
- An immunomodulatory agent may be an immunosuppressive agent.
- immunosuppressive agent it is meant a molecule or compound that reduces (downregulates) the activity and/or efficacy of the immune response, or directs, redirects or reprograms the immune response in a manner that alleviates an undesired result (e.g. an autoimmune response or allergy).
- immunosuppressive agent including calcineurin inhibitors, interleukin inhibitors, selective immunosuppressants and THF-alpha inhibitors.
- An immunomodulatory agent may be an immunosuppressant selected from 5-fluorouracil, 6-thioguanine, adalimumab, anakinra, Atgam, abatacept, alefacept, azathioprine, basiliximab, belatacept, belimumab, benralizumab, brodalumab, canakinumab, certolizumab, chlorambucil, cyclosporine, daclizumab, dimethyl fumerate, dupilumab, eculizumab, efalizumab, ethanercept, everolimus, fingolimod, golimumab, guselkumab, imiquimod, infliximab, ixekizumab, leflunomide, lenlidomide, mechlorethamine, mepolizumab, methotrexate, muromonab-cd3, mycophenolate mofetil,
- An immunomodulatory agent may be any molecule or compound that is an immunosuppressive cytotoxic drug.
- An immunosuppressive cytotoxic drug may include a glucocorticoid, a cytostatic (e.g. alkylating agents, antimetabolites), an antibody, a drug acting on immunophilins, an interferon, an opioid, or a TNF binding protein.
- Immunosuppressive cytotoxic drugs include nitrogen mustards (e.g. cyclophosphamide), nitrosoureas, platinum compounds, folic acid analogs (e.g. methotrexate), purine analogs (e.g. azathioprine and mercaptopurine), pyrimidine analogs (e.g.
- an immunomodulatory agent may be an anti- inflammatory agent.
- An anti-inflammatory agent may be a non-steroidal anti-inflammatory agent (such as a Cox-1 and/or Cox-2 inhibitor), a non-steroidal anti-inflammatory agent, aspirin, salsalate, diflunisal, ibuprofen, fenoprofen, flubiprofen, fenamate, ketoprofen, nabumetone, piroxicam, naproxen, diclofenac, indomethacin, sulindac, tolmetin, etodolac, ketorolac, oxaprozin, a corticosteroid, or celecoxib.
- a non-steroidal anti-inflammatory agent such as a Cox-1 and/or Cox-2 inhibitor
- a non-steroidal anti-inflammatory agent such aspirin, salsalate, diflunisal, ibuprofen, fenoprofen, flubiprofen, fenamate, ketoprofen, nabumetone,
- An immunomodulatory agent may be an anti-rheumatic agent such as prednisone, dexamethasone, chloroquine, hydroxychloroquine, methotrexate, sulfasalazine, cyclosporine, azathioprine, cyclophosphamide, azathioprine, sulfasalazine, penicillamine, aurothioglucose, gold sodium thiomalate, auranofin, methotrexate, mechlorethamine, cyclophosphamide, chlorambucil, or azathioprine.
- an anti-rheumatic agent such as prednisone, dexamethasone, chloroquine, hydroxychloroquine, methotrexate, sulfasalazine, cyclosporine, azathioprine, cyclophosphamide, azathioprine, sulfas
- the therapeutic agent is a polynucleotide encoding a polypeptide, or an interfering polynucleotide (such as miRNA, siRNA shRNA, DsiRNA, or a polynucleotide encoding any thereof).
- a therapeutic agent may be a polynucleotide that contains sequences that correspond largely to the sense or antisense sequence of specific genes or their products, and hence have a direct effect on the expression of these genes and/or their products.
- the use of polynucleotides that contain gene coding sequences affects the transcription and/or translation of the genes of interest in cells that uptake such polynucleotides.
- RNA interference polynucleotides affects the expression of specific genes of interest by directly affecting the levels of mRNA in cells that uptake such nucleotides. This differs significantly from other polynucleotide-based molecules such as CpG, and DNA- or RNA- based poly I:C adjuvants, which do not act through the presence of gene specific sequences.
- an active agent may be a polynucleotide that is not expressed as a protein in a cell, but rather encodes, for example, an antisense RNA, an interfering RNA, a catalytic RNA, or a ribozyme.
- RNA interference is a sequence specific post-transcriptional gene silencing mechanism, which is triggered by double-stranded RNA such as small (or short) interference RNA (siRNA), short hairpin RNA (shRNA), and single stranded intracellular RNA such as microRNA (miRNA), all of which can cause degradation of homologous mRNAs in a cell.
- Interfering RNA may be a naturally occurring or synthetic RNA chain of varying length. Interfering RNA can be duplexes, usually but not always limited to, 20 to 25-nt long that have 19 base pair central double stranded domain with terminal 2-base 3’ overhangs.
- Interfering RNA can be further modified chemically to enhance its in vivo efficacy, induce nuclease-resistance to prevent degradation and enhance stability.
- the anti-sense strand may have either a free 5’-OH or 5’-phosphate terminus, the latter results in natural Dicer processing and represents the active form of the molecule.
- Interfering RNA may have phosphorothioate or boranohosphate modification of the internucleoside linkage to improve nuclease stability and prolong life of the duplex when exposed to serum or other nuclease sources.
- Interfering RNA may have modifications at 2’position, for example, 2’-O-methyl RNA residue incorporation to retain full potency compared with unmodified RNA, retaining stability in serum and significantly reducing the risk of potential IFN responses in the cell. Interfering RNA may also have 2’-fluoro modification, which is usually incorporated selectively at pyrimidine bases, to improve stability and potency.
- the therapeutic agent is a T-helper peptide.
- T-helper peptide refers to a peptide that is an epitope strongly recognized by T cells when displayed on MHC class I/II molecules.
- T-helper peptides are recognized by helper T cells (CD4+ T cells), which play an important role in establishing and maximizing the capabilities of the immune system, and are involved in activating and directing other immune cells, such as for example cytotoxic T cells and B cells. Accordingly, T-helper peptides are capable of enhancing or stimulating an immune response to an antigen.
- Immunodominant T-helper peptides or universal T-helper peptides are known peptides that are broadly reactive in animal and human populations with widely divergent MHC types. T-helper peptides may be comprised in an antigen, or provided with an antigen to boost the immune response to the antigen.
- a T helper peptide may include an immunodominant or universal T helper peptide as known in the art such as, for example but not limited to the modified Tetanus toxin peptide A16L (830-844, AQYIKANSKFIGITEL, with an alanine residue added to its amino terminus to enhance stability); PADRE (pan-DR epitope) comprising the peptide sequence AKXVAAWTLKAAA, wherein X may be phenylalanine or cyclohexylalanine; or Tetanus toxin peptide F21E (947-967, FNNFTVSFWLRVPKVSASHLE).
- an immunodominant or universal T helper peptide as known in the art such as, for example but not limited to the modified Tetanus toxin peptide A16L (830-844, AQYIKANSKFIGITEL, with an alanine residue added to its amino terminus to enhance stability); PADRE (pan-DR epitop
- hydrophobic carrier The pharmaceutical compositions of the present comprise a hydrophobic carrier.
- the hydrophobic carrier may be an essentially pure hydrophobic substance or a mixture of hydrophobic substances. Hydrophobic substances that are useful in the compositions described herein are those that are pharmaceutically acceptable.
- the hydrophobic carrier is typically a liquid but certain hydrophobic carriers that are not liquids at standard room temperature (about 18-25 o C) may be liquefied, for example by warming, and may also be useful.
- the hydrophobic carrier is an oil or a mixture of oils.
- the hydrophobic carrier is a mineral oil (such as Drakeol TM 6VR or IFA), a vegetable oil (such as soybean oil), a nut oil (such as peanut oil), or a mixture of any thereof.
- the hydrophobic carrier is Incomplete Freund’s Adjuvant (IFA), a mineral oil-based hydrophobic carrier.
- the hydrophobic carrier is mannide monooleate in mineral oil, such as commercially available Montanide TM ISA 51 (SEPPIC, France). Montanide TM ISA 51 is a mixture of highly purified mineral oil (Drakeol TM 6VR) and mannide monooleate.
- the hydrophobic carrier is mannide oleate in non- mineral oil, such as commercially available Montanide TM ISA 720 (SEPPIC, France).
- the hydrophobic carrier is MS80 oil which is a mixture of mineral oil (Sigma Aldrich) and a sorbitan monooleate (such as Span TM 80), the components of which can be purchased separately and mixed prior to use.
- the amount of hydrophobic carrier used will depend on the desired volume of the final composition and/or the desired concentration of the components suspended and/or solubilized in the hydrophobic carrier.
- the compositions of the present invention are water-free or substantially free of water, meaning that the compositions are not emulsions.
- water-free it is meant that the compositions contain no water at all.
- the compositions may be substantially free of water.
- substantially free of water is intended to encompass embodiments where the hydrophobic carrier may still contain small quantities of water, provided that the water is present in the non-continuous phase of the carrier.
- individual components of the composition may have small quantities of bound water that may not be completely removed by processes such as lyophilization or evaporation and certain hydrophobic carriers may contain small amounts of water dissolved therein.
- compositions as disclosed herein that are “substantially free of water” contain, for example, less than about 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, 0.1%, 0.05% or 0.01% water on a weight/weight basis of the total weight of the carrier component of the composition.
- the compositions that still contain small quantities of water do not contain a sufficient amount of water such that an emulsion would be formed.
- an “emulsion” refers to a mixture of two or more liquids that are normally immiscible wherein droplets of one liquid are dispersed in the other.
- a hydrophobic substance e.g.
- a dispersion of water droplets in oil is a water-in-oil (W/O) emulsion in which the water (aqueous phase) forms a discontinuous phase and the oil (hydrophobic phase) forms a continuous phase.
- W/O water-in-oil emulsion
- Water-in-oil emulsion or “W/O”, as used herein, refers to an emulsion of a hydrophobic phase in an aqueous phase.
- a dispersion of oil droplets in water is an oil-in-water (O/W) emulsion in which the oil (hydrophobic phase) forms a discontinuous phase and the water (aqueous phase) forms a continuous phase.
- Oil-in-water emulsion or “O/W”, as used herein, refers to an emulsion of a hydrophobic phase in an aqueous phase.
- Conventional emulsion refers to emulsions are composed of numerous emulsifier-coated fluid droplets dispersed within another immiscible fluid medium, wherein the emulsifier forms an interfacial thin layer surrounding individual droplets that create interactions with hydrophilic and hydrophobic phases.
- the pharmaceutical compositions and dried preparations of the present invention comprise at least lipid.
- the lipid is an amphipathic lipid or a mixture of amphipathic lipids.
- any amphipathic lipid may be used, particularly suitable lipids may include those with at least one fatty acid chain containing at least 4 carbons, and typically about 4 to 28 carbons in length.
- the fatty acid chain may contain any number of saturated and/or unsaturated bonds.
- amphipathic lipids which may be used in the pharmaceutical compositions of the present invention, and the pharmaceutical compositions of the present invention may contain a single type of amphipathic lipid or a mixture of different types of amphipathic lipids.
- the amphipathic lipid may be a natural lipid or a synthetic lipid.
- amphipathic lipids for use in the present invention include phospholipids, sphingolipids, sphingomyelin, cerobrocides, gangliosides, ether lipids, sterols, cholesterol, glycerophospholipid, cardiolipin, cationic lipids, anionic lipids, and lipids modified with poly (ethylene glycol) and other polymers.
- Synthetic lipids may include, without limitation, the following fatty acid constituents: lauroyl, myristoyl, palmitoyl, stearoyl, arachidoyl, oleoyl, linoleoyl, erucoyl, or combinations of these fatty acids.
- An “amphipathic lipid” is a lipid having both hydrophilic and hydrophobic parts or characteristics.
- the term “amphipathic” may be used interchangeably with “amphiphile” or “amphiphilic”.
- the hydrophobic portion of an amphipathic lipid may be a large hydrocarbon moiety, either linear or cyclic, such as a long chain of the form CH3(CH2)n, with n > 4.
- the hydrophilic portion of an amphipathic lipid may be either a charged group or a polar uncharged group.
- Charged groups include anionic and cationic groups. Examples of anionic charged groups include the following (wherein the hydrophobic part of the molecule is represented by "R"): carboxylates: RCO 2 ⁇ ; sulfates: RSO 4 ⁇ ; sulfonates: RSO3 ⁇ ; and phosphates (the charged functionality in phospholipids).
- Cationic charged groups include, for example, amines: RNH3 + ("R" again representing the hydrophobic part of the molecule).
- Uncharged polar groups include, for example, alcohols with large R groups, such as diacyl glycerol (DAG).
- Amphipathic lipids may have several hydrophobic parts, several hydrophilic parts, or several of both. Cholesterol is also an amphiphilic lipid.
- the lipid is a phospholipid or a mixture of phospholipids. Broadly defined, a “phospholipid” is a member of a group of lipid compounds that yield on hydrolysis phosphoric acid, an alcohol, fatty acid, and nitrogenous base.
- Phospholipids that may be used include, for example and without limitation, those with at least one head group selected from the group consisting of phosphoglycerol, phosphoethanolamine, phosphoserine, phosphocholine (for example DOPC; 1,2-Dioleoyl-sn-glycero-3-phosphocholine) and phosphoinositol.
- the phospholipid may be phosphatidylcholine or a mixture of lipids comprising phosphatidylcholine.
- the phospholipid may be DOPC or a mixture of phospholipids such as a lecithin (for example Lipoid S100 lecitihin).
- Lecithin is a mixture of phospholipids typically derived from biological sources such as eggs, soybean, and other vegetable sources.
- the phospholipid is sphingomyelin.
- Sphingomyelin contains sphingosine, an amino alcohol with a long unsaturated hydrocarbon chain.
- a fatty acyl side chain is linked to the amino group of sphingosine by an amide bond, to form ceramide.
- the hydroxyl group of sphingosine is esterified to phosphocholine.
- the at least one lipid is present in the pharmaceutical composition at a concentration of 60-160 mg/mL such as, for example 132 mg/mL.
- the at least one lipid is present in the pharmaceutical composition at a concentration of at least 60 mg/mL, at least 70 mg/mL, at least 80 mg/mL, at least 90 mg/mL, at least 100 mg/mL, at least 110 mg/mL, at least 120 mg/mL, at least 132 mg/mL, at least 140 mg/mL, at least 150 mg/mL, or at least 160 mg/mL.
- the at least one lipid is a mixture of a phospholipid and a sterol such as cholesterol.
- the at least one lipid is a mixture of DOPC and cholesterol.
- the at least one lipid is a mixture of lecithin and cholesterol.
- the cholesterol or other sterol is used in an amount equivalent to about 2%, 5%, 10%, 12%, 15%, or 20% of the weight of phospholipid or mixture of phospholipids. In some embodiments, the amount of cholesterol or other sterol used is determined as the amount to sufficiently stabilize the formation of lipid-based structures formed by the phospholipid when suspended in the hydrophobic carrier.
- the term “lipid vesicle particle” may be used interchangeably with “lipid vesicle”.
- a lipid vesicle particle refers to a complex or structure having an internal environment separated from the external environment by a continuous layer of enveloping lipids.
- the expression “layer of enveloping lipids” can mean a single layer lipid membrane (e.g. as found on a micelle or reverse micelle), a bilayer lipid membrane (e.g. as found on a liposome) or any multilayer membrane formed from single and/or bilayer lipid membranes.
- the layer of enveloping lipids is typically a single layer, bilayer or multilayer throughout its circumference, but it is contemplated that other conformations may be possible such that the layer has different configurations over its circumference.
- the lipid vesicle particle may contain, within its internal environment, other vesicle structures (i.e. it may be multivesicular).
- lipid vesicle particle encompasses many different types of structures, including without limitation micelles, reverse micelles, unilamellar liposomes, multilamellar liposomes and multivesicular liposomes.
- the lipid vesicle particles may take on various different shapes, and the shape may change at any given time (e.g. upon drying, sizing, or mixing with therapeutic agents).
- lipid vesicle particles are spherical or substantially spherical structures.
- substantially spherical it is meant that the lipid vesicles are close to spherical, but may not be a perfect sphere.
- lipid vesicle particles include, without limitation, oval, oblong, square, rectangular, triangular, cuboid, crescent, diamond, cylinder or hemisphere shapes. Any regular or irregular shape may be formed.
- a single lipid vesicle particle may comprise different shapes if it is multivesicular.
- the outer vesicle shape may be oblong or rectangular while an inner vesicle may be spherical.
- Exemplary embodiments of lipid vesicle particles include, without limitation, single layer vesicular structures (e.g. micelles or reverse micelles) and bilayer vesicular structures (e.g.
- single layer it is meant that the lipids do not form a bilayer, but rather remain in a layer with the hydrophobic part oriented on one side and the hydrophilic part oriented on the opposite side.
- bilayer it is meant that the lipids form a two-layered sheet, such as with the hydrophobic part of each layer internally oriented toward the center of the bilayer with the hydrophilic part externally oriented.
- the opposite configuration is also possible, i.e. with the hydrophilic part of each layer internally oriented toward the center of the bilayer with the hydrophobic part externally oriented.
- the term “multilayer” is meant to encompass any combination of single and bilayer structures.
- the form adopted may depend upon the specific lipid that is used.
- the lipid vesicle particles may be formed from single layer lipid membranes, bilayer lipid membranes and/or multilayer lipid membranes.
- the lipid membranes are predominantly comprised of and formed by lipids, but may also comprise additional components.
- the lipid membrane may include stabilizing molecules to aid in maintaining the integrity of the structure. Any available stabilizing molecule may be used such as, for example, a sterol such as cholesterol.
- the lipid vesicle particles may be formed by lipids, such as amphipathic lipids, as disclosed herein.
- Lipid vesicle particles may be comprised of a single layer lipid assembly.
- the compositions disclosed herein may comprise a single type of lipid vesicle particle having a single layer lipid assembly or comprise a mixture of different such lipid vesicle particles.
- Lipid vesicle particles having a single layer lipid assembly may comprise aggregates of lipids with the hydrophobic part of the lipids oriented outwards toward the hydrophobic carrier and the hydrophilic part of the lipids aggregating as a core. These structures do not necessarily form a continuous lipid layer membrane.
- Lipid vesicle particles having a single layer lipid assembly comprise reverse micelles.
- Lipid vesicle particles may be bilayer vesicular structures, such as for example, a liposome. Liposomes are completely closed lipid bilayer membranes.
- Liposomes may be unilamellar vesicles (possessing a single bilayer membrane), multilamellar vesicles (characterized by multimembrane bilayers whereby each bilayer may or may not be separated from the next by an aqueous layer) or multivesicular vesicles (possessing one or more vesicles within a vesicle).
- Liposomes form in aqueous or partially aqueous environments, and may form when the compositions herein are not water-free or may form in the aqueous solvents and solutions used in the preparation of pharmaceutical compositions.
- the lipid vesicle particles are liposomes.
- the liposomes are unilamellar, multilamellar, multivesicular or a mixture thereof.
- the mean particle size of the liposomes is ⁇ 80 nm.
- the mean particle size of the liposomes used in the methods disclosed herein is in the range of 80 nm to 120 nm, with a PDI of ⁇ 0.1.
- mean refers to the arithmetic mean of the particle size of the lipid vesicle particles in a given population. It is a synonym for average.
- lipid vesicle particles may not be perfectly spherical, and therefore the “particle size” of a given vesicle particle may not be an exact measure of its diameter.
- the particle size may be defined by other means known in the art, including for example: the diameter of the sphere of equal area or the largest perpendicular distance between parallel tangents touching opposite sides of the particle (Feret’s statistical diameter).
- the sized lipid vesicle particles have a mean particle size of less than or equal to 120 nanometers (i.e. ⁇ 120 nm) and a polydispersity index (PDI) of ⁇ 0.1.
- the sized lipid vesicle particles have a mean particle size of ⁇ 115 nm, more particularly still ⁇ 110 nm and more particularly still ⁇ 100 nm.
- the mean particle size of the sized lipid vesicle particles is between 50 nm and 120 nm. In an embodiment, the mean particle size of the sized lipid vesicle particles is between 80 nm and 120 nm. In an embodiment, the mean particle size of the sized lipid vesicle particles is between about 80 nm and about 115 nm, about 85 nm and about 115 nm, about 90 nm and about 115 nm, about 95 nm and about 115 nm, about 100 nm and about 115 nm or about 105 nm and about 115 nm.
- the mean particle size of the sized lipid vesicle particles is about 80 nm, about 81 nm, about 82 nm, about 83 nm, about 84 nm, about 85 nm, about 86 nm, about 87 nm, about 88 nm, about 89 nm, about 90 nm, about 91 nm, about 92 nm, about 93 nm, about 94 nm, about 95 nm, about 96 nm, about 97 nm, about 98 nm, about 99 nm, about 100 nm, about 101 nm, about 102 nm, about 103 nm, about 104 nm, about 105 nm, about 106 nm, about 107 nm, about 108 nm, about 109 nm, about 110 nm, about 111 nm, about 112 nm, about 113 nm, about 114
- the mean particle size is 120 nm.
- polydispersity index is a measure of the size distribution of the lipid vesicle particles. It is known in the art that the term “polydispersity” may be used interchangeably with “dispersity”.
- the PDI can be calculated by determining the mean particle size of the lipid vesicle particles and the standard deviation from that size. There are techniques and instruments available for measuring the PDI of lipid vesicle particles. For example, DLS is a well-established technique for measuring the particle size and size distribution of particles in the submicron size range, with available technology to measure particle sizes of less than 1 nm (LS Instruments, CH; Malvern Instruments, UK).
- Lipid vesicle particles having a mean particle size of ⁇ 120 nm and a PDI of ⁇ 0.1 may be prepared and provided by any suitable means.
- the lipid vesicle particles are prepared in a manner in which their size is controlled in order to achieve the mean particle size of ⁇ 120 nm and a PDI of ⁇ 0.1.
- lipid vesicle particles are subjected to one or more sizing steps or protocols to achieve the mean particle size of ⁇ 120 nm and a PDI of ⁇ 0.1.
- the lipid vesicle particles may be prepared and provided by any combination of controlling their size during manufacture, performing sizing steps and/or any other means available in the art.
- the lipid vesicle particles must be subjected to one or more active steps of sizing in order to achieve the mean particle size of ⁇ 120 nm and a PDI of ⁇ 0.1.
- the sizing is performed by filter-extrusion whereby lipid vesicle particles are passed through a filter membrane or a series of filter membranes (e.g. polycarbonate membranes) of appropriate pore size.
- sized lipid vesicle particles refers to lipid vesicle particles that have been prepared by a means in which their size is controlled to attain a mean particle size of ⁇ 120 nm and a PDI of ⁇ 0.1 and/or they are sized to meet the criteria of having a mean particle size of ⁇ 120 nm and a PDI of ⁇ 0.1.
- the skilled person will be well aware of techniques available for providing lipid vesicle particles having a mean particle size of ⁇ 120 nm and a PDI of ⁇ 0.1.
- non- sized lipid vesicle particles or a “non-sized lipid vesicle particle preparation” means that the lipid vesicle particles have not be subject to procedures that limit their size to meet the defined size criteria, and/or they do not have a mean particle size of ⁇ 120 nm and a PDI of ⁇ 0.1.
- the sized lipid vesicle particles may be prepared from a lipid precursor that naturally forms lipid vesicle particles of the required size.
- the sized lipid vesicle particles may be prepared using Presome® (Nippon Fine Chemical, Japan).
- Presome® is a dry powder precursor made up of different lipid combinations.
- Presome® is supplied ready to be wetted in a suitable buffer to prepare liposomes.
- the liposomes formed from Presome® have an average particle size of about 93 nm, and sizing procedures (e.g. membrane extrusion, high pressure homogenization, etc.) can be used to achieve the required mean particle size of ⁇ 120 nm and PDI of ⁇ 0.1.
- Presome® may for example be wetted in sodium acetate, pH 9.0 ⁇ 0.5 to form liposomes.
- the Presome® bulk dry powder may be made from DOPC/cholesterol (10:1 (w/w)) or DOPC alone.
- lipid vesicle particles of any size may be employed.
- conventional liposome forming processes may be used, such as the hydration of solvent-solubilized lipids. Exemplary methods of preparing liposomes are discussed, for example, in Gregoriadis 1990; and Frezard 1999.
- the non-sized lipid vesicle particle preparation is subjected to a sizing procedure to obtain lipid vesicle particles having a mean particle size of ⁇ 120 nm and a PDI of ⁇ 0.1.
- a sizing procedure there are various techniques available for sizing lipid vesicle particles (see e.g. Akbarzadeh 2013).
- the non-sized lipid vesicle particle preparation may be sized by high pressure homogenization (microfluidizers), sonication or membrane based extrusion.
- the sizing of the non-sized lipid vesicle particle preparation is performed using high pressure homogenization to obtain sized lipid vesicle particles having a mean particle size of >120 nm and a PDI of >0.1, and the lipid vesicle particles obtained via high pressure homogenization can then be further sized down using membrane based extrusion.
- membrane based extrusion is performed by passing the sized lipid vesicle particle preparation 5-20 times through a 0.1 um polycarbonate membrane or, alternatively, 5-20 times through a 0.08 um polycarbonate membrane, thereby attaining a mean particle size of ⁇ 120 nm and PDI of ⁇ 0.1.
- the sized lipid vesicle particles may be prepared by adding the lipids to a suitable solvent (e.g. sodium phosphate, 50 mM, pH 7.0), shaking and/or stirring the lipid mixture (e.g. at 300 RPM for about 1 hour) and using membrane based extrusion to obtain the sized lipid vesicle particles.
- a suitable solvent e.g. sodium phosphate, 50 mM, pH 7.0
- shaking and/or stirring the lipid mixture e.g. at 300 RPM for about 1 hour
- Exemplary, non-limiting embodiments of membrane based extrusion include: (i) passing a non-sized lipid vesicle particle preparation 20-40 times through a 0.2 ⁇ m polycarbonate membrane, and then 10-20 times through a 0.1 ⁇ m polycarbonate membrane; or (ii) passing a non-sized lipid vesicle particle preparation 20-40 times through a 0.2 ⁇ m polycarbonate membrane, then 10-20 times through a 0.1 ⁇ m polycarbonate membrane, and then 10-20 times through a 0.08 ⁇ m polycarbonate membrane.
- the sizing may be performed by passing a non-sized lipid vesicle particle preparation 25 times through a 0.2 ⁇ m polycarbonate membrane, and then 10 times through a 0.1 ⁇ m polycarbonate membrane.
- the sizing may be performed by passing a non-sized lipid vesicle particle preparation 25 times through a 0.2 ⁇ m polycarbonate membrane, then 10 times through a 0.1 ⁇ m polycarbonate membrane, and then 15 times through a 0.08 ⁇ m polycarbonate membrane.
- the term “dried preparation” refers to a mixture of components as described herein that is dried by a technique known in the art in order to remove all or substantially all of a solvent.
- the term “dried preparation” does not necessarily mean that the preparation is completely dry. For example, depending on the solvent or solvents used in the methods disclosed herein, it is possible that a small component of volatile and/or non-volatile material will remain in the dried preparation. In an embodiment, the non-volatile material will remain.
- dried preparation it is meant that the preparation no longer contains substantial quantities of water and/or organic solvent. The process used to dry the preparation should be capable of removing substantially all water and/or organic solvent.
- the dried preparation is completely free of water and/or organic solvent.
- the dried preparation may contain a residual moisture content based on the limitations of the drying process (e.g. lyophilization). This residual moisture content will typically be less than 2%, less than 1%, less than 0.5%, less than 0.25%, less than 0.1%, less than 0.05% or less by weight of the dried preparation. This residual moisture content will not be more than 5% by weight of the dried preparation as this would result in a product that is not clear.
- Various methods may be used to dry compositions are known in the art. In an embodiment, the drying is performed by lyophilization, spray freeze-drying, or spray drying.
- the drying is performed by lyophilization.
- lyophilization works by freezing the material and then reducing the surrounding pressure to allow the volatile solvent (e.g. water) in the material to sublime directly from the solid phase to the gas phase. Any conventional freeze-drying procedure may be used to carry out the drying step of the methods disclosed herein.
- the lyophilization is performed by sequential steps of loading, freezing, evacuation and drying (e.g. primary drying and secondary drying).
- lyophilization of the sized lipid vesicle particle/therapeutic agent mixture can be performed within a sealed bag in a benchtop freeze dryer.
- This may be particularly advantageous because it reduces the number of steps that must be performed in a sterile laboratory environment and allows for the rapid manufacture of smaller batch sizes.
- aseptically filled vials containing the mixture can be loaded and sealed within a sterile bag under sterile conditions.
- These sterile, sealed units can then undergo lyophilization in an open laboratory (i.e. non-sterile environment) using a benchtop freeze dryer.
- the freeze dryer is a benchtop freeze dryer.
- the freeze dryer is a Virtis benchtop lyophilizer.
- the freeze dryer is in an open laboratory (i.e. non-sterile environment).
- Sterilization may be performed by any method known in the art.
- the sterilization is performed by sterile filtration, steam heat sterilization, irradiation (e.g. gamma irradiation) or chemical sterilization.
- the sterilization is performed by sterile filtration.
- the sterile filtration is be performed before drying. Any conventional procedure for sterile filtration may be employed so long as it does not affect the solubility and stability of the therapeutic agents, lipid-based adjuvant, and/or polyI:C polynucleotide in the sized lipid vesicle particle/therapeutic agent mixture.
- it may be desirable to perform the sterile filtration under low pressure conditions e.g.
- the serial filtration may be performed using commercially available sterile filtration membranes (e.g. MilliporeSigma). In an embodiment, the sterile filtration is performed using a 0.22 ⁇ m-rated membrane, a 0.2 ⁇ m-rated membrane and/or a 0.1 ⁇ m-rated membrane. In an embodiment, the sterile filtration is performed by a single passage of the mixture through a single filtration membrane. In another embodiment, the sterile filtration is performed by serially passing the mixture sequentially through a combination of the same or different sized filtration membranes.
- sterile filtration membranes e.g. MilliporeSigma
- the methods disclosed herein may further comprise a step of confirming that the sized lipid vesicle particles have retained a mean particle size of ⁇ 120 nm and PDI of ⁇ 0.1.
- a step of confirming that the sized lipid vesicle particles have retained a mean particle size of ⁇ 120 nm and PDI of ⁇ 0.1 there are several techniques, instruments and services that are available to measure the mean particle size and PDI of lipid vesicle particles, such as for example and without limitation TEM, SEM, AFM, FTIR, XPS, XRD, MALDI-TOF-MS, NMR and DLS.
- the step of confirming the size and PDI of the lipid vesicle particles is performed using a DLS ZETASIZER NANO-S particle size analyzer.
- the methods disclosed herein may further comprise a step of evaluating the stability of the lipids, therapeutic agent(s), lipid-based adjuvant, and/or polyI:C polynucleotide before and/or after the drying step.
- the stability of the components may be measured by any known means or method.
- stability of the dried preparation may be determined by the appearance of the dried preparation (lyophilisate) or measurement of the content of the components over time (e.g. by HPLC, RP-HPLC, IEX-HPLC, etc.).
- HPLC is a technique which can be used to separate, identify and quantify each component in a mixture.
- stability may evaluated upon solubilization in a hydrophobic carrier by various methods, such as for example: appearance of the solubilized product; identification and quantification of lipids, therapeutic agent(s), lipid-based adjuvant, and/or polyI:C polynucleotide, impurities or degradants (e.g. by RP-HPLC, IEX-HPLC, etc.); particle size of the lipid vesicle particles (e.g.
- lipids may first be dissolved and mixed in an organic solvent. In embodiments where different types of lipid are used, this step will allow a homogenous mixture of the lipids to be formed. In an embodiment, these steps may be carried out in chloroform, chloroform:methanol mixtures, tertiary butanol or cyclohexane.
- the lipids are prepared at 10-20mg lipid/mL organic solvent; however, higher or lower concentrations may also be used.
- the organic solvent is removed (e.g. by evaporation) to yield a lipid film.
- the lipid film may then be frozen and lyophilized to yield a dry lipid film.
- the dry lipid film may then be hydrated with an aqueous solution containing therapeutic agents/polyI:C polynucleotide/lipid-based adjuvant to provide a non-sized lipid vesicle particle preparation.
- the step of hydration may be performed with shaking and/or mixing (e.g. at 300 RPM for about 1 hour).
- an aqueous solution of lipids may be combined with at least one solution containing therapeutic agents/polyI:C polynucleotide/lipid-based adjuvant.
- one or more dry therapeutic agents/polyI:C polynucleotide/lipid-based adjuvant may be added to, and solubilized in, the aqueous solution of lipids or sized lipid vesicle preparation. These embodiments may be performed with shaking and/or mixing (e.g. at 300 RPM for about 1 hour).
- therapeutic agents/polyI:C polynucleotide/lipid-based adjuvant are either solubilized in a solvent (e.g. aqueous or organic) prior to mixing with lipid vesicle particles or therapeutic agents/polyI:C polynucleotide/lipid-based adjuvant are solubilized upon being mixed with the lipid vesicle particles.
- a solvent e.g. aqueous or organic
- therapeutic agents/polyI:C polynucleotide/lipid-based adjuvant may be added as a dry powder to a solution containing lipid vesicle particles or both the lipid vesicle particles and therapeutic agents/polyI:C polynucleotide/lipid-based adjuvant may be mixed together in a fresh solvent.
- the pharmaceutical compositions disclosed herein may find application in any instance in which it is desired to administer therapeutic agents to a subject.
- the subject may be a vertebrate, such as a fish, bird or mammal.
- the subject is a mammal.
- the subject is a human.
- the pharmaceutical compositions may be used in methods for treating, preventing or diagnosing a disease, disorder or condition to which the therapeutic agent is targeted.
- the pharmaceutical compositions may be used in methods for delivering a therapeutic agent to a subject.
- the methods comprise administering to a subject the pharmaceutical composition as described herein.
- the pharmaceutical compositions may be used in methods for modulating an immune response in a subject.
- modulating is intended to refer to both immunostimulation (e.g. inducing or enhancing an immune response) and immunosuppression (e.g. preventing or decreasing an immune response).
- the method would involve one or the other of immunostimulation or immunosuppression, but it is possible that the method could be directed to both.
- the “immune response” may either be a cell-mediated (CTL) immune response or an antibody (humoral) immune response.
- the pharmaceutical compositions disclosed herein may be used for inducing a cell-mediated immune response to the therapeutic agents (e.g. antigens).
- to “induce” an immune response is to elicit and/or potentiate an immune response.
- the pharmaceutical compositions disclosed herein may be used for inducing an antibody immune response to the therapeutic agents (e.g. antigens).
- an “antibody immune response” or “humoral immune response” (used interchangeably herein), as opposed to cell-mediated immunity, is mediated by secreted antibodies which are produced in the cells of the B lymphocyte lineage (B cells).
- B cells B lymphocyte lineage
- Such secreted antibodies bind to antigens, such as for example those on the surfaces of foreign substances, pathogens (e.g.
- the pharmaceutical compositions disclosed herein may be useful for treating or preventing diseases and/or disorders ameliorated by a cell-mediated immune response or a humoral immune response.
- the pharmaceutical compositions disclosed herein may find application in any instance in which it is desired to administer therapeutic agents (e.g. antigens) to a subject to induce a cell-mediated immune response or a humoral immune response.
- the pharmaceutical compositions is for inducing an antibody immune response and/or cell-mediated immune response to the therapeutic agents (e.g. antigens) in said subject.
- pharmaceutical compositions is for the treatment and/or prevention of an infectious disease or cancer.
- Treating” or “treatment of”, or “preventing” or “prevention of”, as used herein, refers to an approach for obtaining beneficial or desired results.
- Beneficial or desired results can include, but are not limited to, alleviation or amelioration of one or more symptoms or conditions, diminishment of extent of disease, stabilisation of the state of disease, prevention of development of disease, prevention of spread of disease, delay or slowing of disease progression (e.g. suppression), delay or slowing of disease onset, conferring protective immunity against a disease-causing agent and amelioration or palliation of the disease state.
- Treating can be distinguished from “preventing” in that “treating” typically occurs in a subject who already has a disease or disorder, or is known to have already been exposed to an infectious agent, whereas “preventing” typically occurs in a subject who does not have a disease or disorder, or is not known to have been exposed to an infectious agent. As will be appreciated, there may be overlap in treatment and prevention.
- the pharmaceutical compositions disclosed herein may be used for treating and/or preventing an infectious disease, such as caused by a viral infection, in a subject in need thereof.
- the subject may be infected with a virus or may be at risk of developing a viral infection.
- Viral infections that may be treated and/or prevented by the use or administration of a pharmaceutical composition as disclosed herein, without limitation, Cowpoxvirus, Vaccinia virus, Pseudocowpox virus, Human herpesvirus 1 , Human herpesvirus 2, Cytomegalovirus, Human adenovirus A-F, Polyomavirus, Human papillomavirus (HPV), Parvovirus, Hepatitis A virus, Hepatitis B virus, Hepatitis C virus, Human immunodeficiency virus, Orthoreovirus, Rotavirus, Ebola virus, parainfluenza virus, influenza A virus, influenza B virus, influenza C virus, Measles virus, Mumps virus, Rubella virus, Pneumovirus, respiratory syncytial virus (RSV), Rabies virus, California encephalitis virus, Japanese encephalitis virus, Hantaan virus, Lymphocytic chor
- exemplary bacterial pathogens may include Anthrax (Bacillus anthracis), Brucella, Bordetella pertussis, Candida, Chlamydia pneumoniae, Chlamydia psittaci, Cholera, Clostridium botulinum, Coccidioides immitis, Cryptococcus, Diphtheria, Escherichia coli O157: H7, Enterohemorrhagic Escherichia coli, Enterotoxigenic Escherichia coli, Haemophilus influenzae, Helicobacter pylori, Legionella, Leptospira, Listeria, Meningococcus, Mycoplasma pneumoniae, Mycobacterium, Pertussis, Pneumonia, Salmonella, Shigella, Staphylococcus, Streptococcus pneumoniae and Yersinia enterocolitica.
- Anthrax Bacillus anthracis
- Brucella Bacillus anthracis
- the bacterial infection is Anthrax.
- exemplary protozoan pathogens may include those of the genus Plasmodium (Plasmodium falciparum, Plasmodium malariae, Plasmodium vivax, Plasmodium ovale or Plasmodium knowlesi), which cause malaria.
- the pharmaceutical compositions disclosed herein may be for use in treating and/or preventing cancer in a subject in need thereof. The subject may have cancer or may be at risk of developing cancer.
- cancer refers to cells that exhibit abnormal growth, characterized by a significant loss of control of cell proliferation or cells that have been immortalized.
- cancer or “tumor” includes metastatic as well as non-metastatic cancer or tumors.
- a cancer may be diagnosed using criteria generally accepted in the art, including the presence of a malignant tumor.
- cancers that may be capable of being treated and/or prevented by the use or administration of a pharmaceutical composition as disclosed herein include carcinoma, adenocarcinoma, lymphoma, leukemia, sarcoma, blastoma, myeloma, and germ cell tumors.
- particularly suitable embodiments may include glioblastoma, multiple myeloma, ovarian cancer, breast cancer, fallopian tube cancer, prostate cancer or peritoneal cancer.
- the cancer may be caused by a pathogen, such as a virus.
- Viruses linked to the development of cancer are known to the skilled person and include, but are not limited to, human papillomaviruses (HPV), John Cunningham virus (JCV), Human herpes virus 8, Epstein Barr Virus (EBV), Merkel cell polyomavirus, Hepatitis C Virus and Human T cell leukaemia virus-1.
- the cancer is one that expresses one or more tumor- specific neoantigens.
- the cancer is breast cancer, ovarian cancer, prostate cancer, fallopian tube cancer, peritoneal cancer, glioblastoma or diffuse large B cell lymphoma.
- the pharmaceutical compositions disclosed herein may be useful for either the treatment or prophylaxis of cancer; for example, a reduction of the severity of cancer (e.g. size of the tumor, aggressiveness and/or invasiveness, malignancy, etc.) or the prevention of cancer recurrences.
- the pharmaceutical composition as disclosed herein may be administered by any suitable route.
- the route of administration is injection, such as subcutaneous injection.
- kits The pharmaceutical compositions disclosed herein are optionally provided to a user as a kit comprising the individual components which may be assembled to produce the pharmaceutical composition.
- the kit is for preparing a composition for the treatment, prevention and/or diagnosis of a disease, disorder or condition or for the delivery of a therapeutic agent.
- the kits can further comprise one or more additional reagents, packaging materials, and an instruction set or user manual detailing preferred methods of using the kit components.
- the containers are vials.
- the invention is further illustrated by the following non-limiting examples.
- Example 1 [0118] First, a vial containing DPX-FP (IMV-Inc, Dartmouth, Canada) was reconstituted in 0.45 mL of Montanide ISA 51 oil diluent (SEPPIC, France) by soaking for 5 minutes and shaking for 2 minuntes, then vortexing for one minute.
- SEPPIC Montanide ISA 51 oil diluent
- DPX-PAM3CSK4 IMV-Inc, Dartmouth, Canada
- DPX-PAM3CSK4 IMV-Inc, Dartmouth, Canada
- reconstitution was completed by soaking for 5 minutes, shaking for 2 minutes, and vortexing for one minute to obtain the final concentrations of: FP peptide 9 ug/50 uL dose; DNA based polyI:C polynucleotide (dIdC) 18 ug/50 uL dose; PAM3CSK42 ug/50 uL dose; and Lipid 12.5 mg/50 uL dose.
- dIdC DNA based polyI:C polynucleotide
- the DPX-FP product vial was previously prepared by adding FP (NeoMPS) and DNA based polyI:C polynucleotide (dIdC) stock (Biospring) to a lipid-mixture solution, mixing well and freeze-drying.
- FP NeoMPS
- dIdC DNA based polyI:C polynucleotide
- Biospring DNA based polyI:C polynucleotide
- a lipid-mixture 132 mg/mL) containing DOPC and cholesterol in a 10:1 ratio (w:w) (Lipoid GmBH, Germany) was dissolved in 40% tertiary-butanol by shaking well at 300 RPM at room temperature for 1 hour or until dissolved.
- FP stock (10 mg/mL) was prepared in DMSO and DNA based polyI:C polynucleotide adjuvant stock (10 mg/mL) was prepared in sterile water.
- DNA based polyI:C polynucleotide adjuvant stock 10 mg/mL was prepared in sterile water.
- 10 ⁇ L of FP stock was added to obtain 0.1 mg/mL final fill concentration, shaken well at 300 RPM for 5 minutes.
- 20 ⁇ L of DNA based polyI:C polynucleotide (dIdC) stock was added to obtain 0.2 mg/mL final fill concentration, shaken well at 300 RPM for 5 minutes.
- the volume was adjusted to 1.0 mL with 40% tertiary-butanol, freeze-dried, and stored at -20 C.
- the dried preparation is reconstituted in 0.45 mL oil diluent when needed.
- the DPX-PAM3CSK4 vial was prepared as follows: Briefly, a homogenous lipid- mixture of DOPC and cholesterol (Lipoid GmbH, Germany) was weighed to obtain 132 mg/mL of the lipid-mixture and was added to sodium phosphate, 100 mM, pH 6.0 with shaking at 300 RPM for about 1 hour.
- the mixture was then sized by passing the material 25 times through a 0.2 ⁇ m polycarbonate membrane, then 10 times through a 0.1 ⁇ m polycarbonate membrane, and then 15 times through a 0.08 ⁇ m polycarbonate membrane to attain a particle size of ⁇ 100 nm with a polydispersity index (PDI) of ⁇ 0.1.
- the lipopeptide adjuvant PAM 3 CSK4 Polypeptide group, San Diego, USA
- the mixture was then sterile filtered, aseptically filled in 1.0 mL aliquots into sterile glass vials, freeze-dried, and stored at -20°C.
- Example 2 [0122] First, a vial containing DPX-MVP-S (IMV-Inc, Dartmouth, Canada) was reconstituted in 0.7 mL of Montanide ISA 51 oil (SEPPIC, France) by soaking for 5 minutes, shaking for 2 mins, and vortexing for one minute.
- DPX-MVP-S IMV-Inc, Dartmouth, Canada
- DPX-PAM3CSK4 IMV-Inc, Dartmouth, Canada
- Restitution was completed by soaking for 5 minutes, shaking for 2 minutes, and vortexing for one minute to obtain the final concentrations of: MVP-S peptide each at 45 ug/50 uL dose; T-helper peptide A16L at 22.5 ug/50 uL dose; DNA based polyI:C polynucleotide (dIdC) 18 ug/50 uL dose; PAM 3 CSK42 ug/50 uL dose; and Lipid 12.5 mg/50 uL dose.
- dIdC DNA based polyI:C polynucleotide
- the DPX-MVP-S vial was prepared as follows: Briefly, DNA based polyI:C polynucleotide (dIdC) (BioSpring GmbH (Frankfurt, Germany) and peptide antigens (PolyPeptide Laboratories (San Diego, CA, USA) stock solutions were prepared as listed below: Stock# Component Solvent 5) in the following order: (4), (2), (3), (5) and then (1). The pH was adjusted to 10.0 ⁇ 0.5.
- a 10:1 (w:w), homogenous lipid-mixture of DOPC and cholesterol (Lipoid GmbH, Germany) was weighed to obtain 132 mg/mL of the lipid-mixture and added to the peptide/polynucleotide solution to form an intermediate bulk (non-sized) and mixed using a Silverson high speed mixer.
- the pH was adjusted to 10.0 ⁇ 0.5, if required.
- the intermediate bulk was then sized using an Emulsiflex C5 or C55 extruder by passing the material 35 times through a 0.2 ⁇ m polycarbonate membrane and then 10 times through a 0.1 ⁇ m polycarbonate membrane to attain a particle size of ⁇ 120 nm with a PDI of ⁇ 0.1.
- the peptide stock solutions (6) and (7) were then added to the sized lipid vesicle particle bulk immediately after preparation.
- the final pH of the solution was adjusted to 7.0 ⁇ 0.5.
- the final preparation was then sterile filtered, aseptically filled (1.6 mL aliquots) into sterile glass vials, freeze-dried, and stored at -20°C.
- the dried preparation is reconstituted in 0.7 mL oil diluent when needed.
- the DPX-PAM3CSK4 vial was prepared as follows: Briefly, homogenous lipid- mixture of DOPC and cholesterol (Lipoid GmbH, Germany) was weighed to obtain 132 mg/mL of the lipid-mixture was added to sodium phosphate, 100 mM, pH 6.0 with shaking at 300 RPM for about 1 hour. The mixture was then sized by passing the material 25 times through a 0.2 ⁇ m polycarbonate membrane, then 10 times through a 0.1 ⁇ m polycarbonate membrane, and then 15 times through a 0.08 ⁇ m polycarbonate membrane to attain a particle size of ⁇ 100 nm with a PDI of ⁇ 0.1.
- the lipopeptide adjuvant PAM3CSK4 (Polypeptide group, San Diego, USA) was then added at 0.02 mg/mL to the sized lipid vesicles with shaking at 300 RPM for about 15 minutes. The mixture was then sterile filtered, aseptically filled to 1.0 mL aliquots into sterile glass vials, freeze-dried, and stored at -20°C. The dried preparation is reconstituted in 0.45 mL oil diluent when needed.
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- Veterinary Medicine (AREA)
- Public Health (AREA)
- General Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Epidemiology (AREA)
- Medicinal Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Engineering & Computer Science (AREA)
- Optics & Photonics (AREA)
- Nanotechnology (AREA)
- Physics & Mathematics (AREA)
- Biomedical Technology (AREA)
- Dispersion Chemistry (AREA)
- Dermatology (AREA)
- Medicinal Preparation (AREA)
Abstract
The present application relates to methods for preparing pharmaceutical compositions comprising therapeutic agents, lipids, a lipid-based adjuvant, and a polyI:C polynucleotide.
Description
METHODS OF MAKING DRIED PHARMACEUTICAL COMPOSITIONS CROSS-REFERENCE TO RELATED APPLICATIONS [0001] This application claims the benefit of U.S. Provisional Application No. 63/449,779 filed March 3, 2023, which is incorporated by reference herein. FIELD [0002] The present application relates to methods for preparing pharmaceutical compositions comprising therapeutic agents, lipids, a lipid-based adjuvant, and a polyI:C polynucleotide. BACKGROUND [0003] In the pharmaceutical field, the effective delivery of therapeutic agents often poses difficulties and challenges, particularly in respect of the complexities of emerging delivery platforms designed to enhance the efficacy of therapeutic agents. For these specialized delivery platforms that employ unique components, new hurdles arise that do not exist for conventional pharmaceutical compositions. This is certainly the case for delivery platforms using water-free, hydrophobic carriers. These challenges are compounded when it is desirable to formulate pharmaceutical compositions comprising multiple components with diverse properties. [0004] In the case of pharmaceutical compositions comprising antigens (i.e. vaccine compositions), vaccines containing antigens are typically not immunogenic enough to generate rapid and prolonged immunity. This can sometimes be overcome with the use of an adjuvant to boost the immune response towards an antigen. There are generally two broad categories of adjuvants: delivery systems and immune-stimulants. The delivery system of a vaccine can act as an adjuvant by providing stability and prolonged interaction of the antigen with the immune system. Vaccine compositions may also incorporate molecular compounds with immune- stimulatory activity as adjuvants with the aim of further enhancing immunogenicity of the vaccine by directly activating cells of the immune system. [0005] Immune-stimulant adjuvants are defined molecular agonists that are recognized by the immune system via specialized receptors, for example polyI:C polynucleotide and lipid- based adjuvants such as PAM3CSK4 stimulate distinct Toll-like receptors. Immune-stimulants can activate the immune system and also direct the type of immune response generated towards a vaccine antigen. For example, the effectiveness of many vaccines is correlated to the generation of antibodies; however, for other types of vaccines a strong cytotoxic immune response primarily
mediated by CD8+ T cells is desired. The type of immune response generated towards a vaccine antigen can be manipulated by including immune-stimulants that activate particular receptors found on immune cells that can initiate these responses through generation of cytokines and chemokines. [0006] Differences in the properties of immune-stimulants such as polyI:C polynucleotide and lipid-based adjuvants can complicate their incorporation into pharmaceutical compositions. SUMMARY [0007] In an embodiment, there is provided a method for preparing a pharmaceutical composition, comprising: (a) providing: (i) a first dried preparation comprising at least one therapeutic agent, a lipid, and a polyI:C polynucleotide, and (ii) a second dried preparation comprising a lipid-based adjuvant and a lipid; (b) combining the first dried preparation with a hydrophobic carrier to produce a reconstituted preparation; and (c) combining the reconstituted preparation with the second dried preparation to produce a pharmaceutical composition. [0008] In an embodiment, there is provided a method for preparing a pharmaceutical composition, comprising: (a) providing: (i) a first dried preparation comprising at least one therapeutic agent, a lipid, and a polyI:C polynucleotide, and (ii) a second dried preparation comprising a lipid-based adjuvant and a lipid; (b) combining the second dried preparation with a hydrophobic carrier to produce a reconstituted preparation; and (c) combining the reconstituted preparation with the first dried preparation to produce a pharmaceutical composition. [0009] In an embodiment, there is provided a pharmaceutical composition prepared by the method as described herein. [0010] In an embodiment, there is provided a method of inducing an antibody and/or CTL immune response in a subject comprising administering to the subject the pharmaceutical composition as described herein. [0011] In an embodiment, there is provided a use of the pharmaceutical composition as described herein for inducing an antibody and/or CTL immune response in a subject. [0012] In an embodiment, there is provided a method of delivering a therapeutic agent to a subject comprising administering to the subject the pharmaceutical composition as described herein.
[0013] In an embodiment, there is provided a use of the pharmaceutical composition as described herein for delivering a therapeutic agent to a subject. [0014] In an embodiment, there is provided a kit for preparing the pharmaceutical composition as described herein, the kit comprising: a container comprising a first dried preparation prepared by the method as described herein; a container comprising second dried preparation prepared by the method as described herein; and a container comprising a hydrophobic carrier. [0015] Other aspects and features of the present invention will become apparent to those of ordinary skill in the art upon review of the following description in conjunction with the accompanying figures. DETAILED DESCRIPTION [0016] Poly I:C polynucleotide is an oligonucleotide molecule, composed of either DNA or RNA nucleosides, which interacts with Toll-like receptors (TLRs) that can detect nucleic acids, such as TLR3. Lipid-based adjuvants such as PAM2Cys and PAM3Cys interact with TLR1/2. Stimulating multiple TLR receptors at once has been reported to have an additive or synergistic activating effect on dendritic cells. Therefore, combining poly I:C polynucleotide and a lipid-based adjuvant such as PAM2Cys or PAM3Cys in a pharmaceutical composition that can ensure they engage with TLRs on the same cell can provide a potent activating effect to the immune system. Optimal doses of each adjuvant may need to be confirmed for different applications. For example, vaccines for cancer therapy may need stronger immune stimulation than vaccines for prevention of infectious disease. [0017] The lipopeptides PAM2Cys and PAM3Cys comprise a lipid palmitic acid moiety conjugated to a cysteine (Cys) residue that may be extended into a peptide with polar residues such as serine (Ser) and/or lysine (Lys) with a positive surface charge. By contrast, polyI:C polynucleotides are made up of single or double stranded molecules containing inosinic acid residues (I) and cytidylic acid residues (C) with a negative surface charge. If needed, these two distinct immune stimulators can be complexed together by electrostatic interactions to a certain extent with concentration restrictions. However, increasing the concentration of any one of these immune stimulators may lead to a pH shift and cause unwanted precipitation during formulation and/or in the finished product, constraining the ability to provide desired concentrations of lipid- based adjuvant and polyI:C polynucleotide in a pharmaceutical composition. Using current
methods, it is only possible to combine polyI:C polynucleotides and PAM2Cys and PAM3Cys lipid-based adjuvants in a single water-free formulation up to about 0.5 mg/mL concentration. [0018] Embodiments of the present invention provide methods of fully incorporating both polyI:C polynucleotide and lipid-based adjuvant (such as PAM2Cys and PAM3Cys) along with therapeutic agents that overcomes concentration limitations, irrespective of the nature of therapeutic agents and formulation buffer/pH used to prepare the pharmaceutical composition. Using the methods of the present invention, higher concentrations of polyI:C polynucleotide and/or lipid-based adjuvant can successfully be incorporated into pharmaceutical compositions. [0019] PolyI:C polynucleotide [0020] PolyI:C polynucleotides are polynucleotide molecules (either RNA or DNA or a combination of DNA and RNA) containing inosinic acid residues (I) and cytidylic acid residues (C), and which induce the production of inflammatory cytokines, such as interferon. In some embodiments, the polyI:C polynucleotide is double-stranded. In double-stranded embodiments, they are typically composed of one strand consisting entirely of cytosine-containing nucleotides and one strand consisting entirely of inosine-containing nucleotides, although other configurations are possible. For instance, each strand may contain both cytosine-containing and inosine-containing nucleotides. In some embodiments, the polyI:C polynucleotide is single- stranded. In single-stranded embodiments, the strand comprises both cytosine-containing and inosine-containing nucleotides. In some instances, either or both strand may additionally contain one or more non-cytosine or non-inosine nucleotides. [0021] In some embodiments, the polyI:C polynucleotide may be a single-stranded molecule containing inosinic acid residues (I) and cytidylic acid residues (C). As an example, and without limitation, the single-stranded polyI:C polynucleotide may be a sequence of repeating dIdC. In a particular embodiment, the sequence of the single-stranded polyI:C may be a 26-mer sequence of (IC)13, i.e. ICICICICICICICICICICICICIC. As the skilled person will appreciate, due to their nature (e.g. complementarity), it is anticipated that these single-stranded molecules of repeating IC or dIdC may form homodimers. [0022] Accordingly, as used herein, a “polyI:C” or “polyI:C polynucleotide” is a double- or single-stranded polynucleotide molecule (RNA or DNA or a combination of DNA and RNA), each strand of which contains at least 6 contiguous inosinic or cytidylic acid residues, or 6 contiguous residues selected from inosinic acid and cytidylic acid in any order (e.g. IICIIC or ICICIC). PolyI:C polynucleotides will typically have a length of about 6, 8, 10, 12, 14, 16, 18,
20, 22, 24, 25, 26, 28, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 150, 200, 250, 300, 500, 1000 or more nucleotides. [0023] Each strand of a double-stranded polyI:C polynucleotide may be a homopolymer of inosinic or cytidylic acid residues, or each strand may be a heteropolymer containing both inosinic and cytidylic acid residues. In either case, the polymer may be interrupted by one or more non-inosinic or non-cytidylic acid residues (e.g. uridine), provided there is at least one contiguous region of 6 I, 6 C or 6 I/C residues as described above. Typically, each strand of a polyI:C polynucleotide will contain no more than 1 non-I/C residue per 6 I/C residues, more preferably, no more than 1 non-I/C residue per every 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 or 30 I/C residues. [0024] The inosinic acid or cytidylic acid (or other) residues in the polyI:C polynucleotide may be derivatized or modified as is known in the art, provided the ability of the polyI:C polynucleotide to promote the production of an inflammatory cytokine, such as interferon, is retained. Non-limiting examples of derivatives or modifications include e.g. azido modifications, fluoro modifications, or the use of thioester (or similar) linkages instead of natural phosphodiester linkages to enhance stability in vivo. The polyI:C polynucleotide may also be modified to e.g. enhance its resistance to degradation in vivo by e.g. complexing the molecule with positively charged poly-lysine and carboxymethylcellulose, or with a positively charged synthetic peptide. [0025] Lipid-based adjuvant [0026] As used herein, a lipid-based adjuvant is an adjuvant that comprises at least one lipid moiety or lipid component. [0027] The expression “lipid moiety” or “lipid component” refers to any fatty acid (e.g. fatty acyls) or derivative thereof, including for example triglycerides, diglycerides, and monoglycerides. Exemplary fatty acids include, without limitation, palmitoyl, myristoyl, stearoyl and decanoyl groups or any C2 to C30 saturated or unsaturated fatty acyl group, preferably any C14 to C22 saturated or unsaturated fatty acyl group, and more preferably a C16 saturated or unsaturated fatty acyl group. Thus, as referred to herein, the expression “lipid-based adjuvant” encompasses adjuvants comprising a fatty acyl group or derivative thereof. [0028] Lipid-based adjuvants contain at a minimum at least one lipid moiety, or a synthetic/semi-synthetic lipid moiety analogue, which can be coupled onto an amino acid, an oligopeptide or other molecules (e.g. a carbohydrate, a glycan, a polysaccharide, biotin,
Rhodamine, etc.). Thus, without limitation, the lipid-based adjuvant may be, for example, a lipoamino acid, a lipopeptide, a lipoglycan, a lipopolysaccharide or a lipoteichoic acid. Moreover, a lipid moiety or a structure containing a lipid moiety can be coupled covalently or non-covalently to an antigen to create antigenic compounds with built-in adjuvanting properties. For example, and without limitation, the lipid-based moiety may comprise a cation (e.g. nickel) to provide a positive charge for non-covalent coupling. [0029] In some embodiments, the lipid moiety or lipid component may be naturally occurring, such as for example a cell-wall component (e.g. lipoprotein) from a Gram-positive or Gram-negative bacteria, Rhodopseudomonas viridis, or mycoplasma. In other embodiments, the lipid moiety or lipid component may be synthetic or semi-synthetic [0030] The lipid-based adjuvant may comprise palmitic acid (PAM) as at least one of the lipid moieties or components of the adjuvant. Such lipid-based adjuvants are referred to herein as a “palmitic acid adjuvant”. Palmitic acid is a low molecular weight lipid found in the immunologically reactive Braun’s lipoprotein of Escherichia coli. Other common chemical names for palmitic acid include, for example, hexadecanoic acid in IUPAC nomenclature and 1-Pentadecanecarboxylic acid. The molecular formula of palmitic acid is CH3(CH2)14CO2H. As will be understood to those skilled in the art, it is possible that the lipid chain of palmitic acid may be altered. Exemplary compounds which may be used herein as palmitic acid adjuvants, and methods for their synthesis, are described for example in United States Patent Publications US 2008/0233143; US 2010/0129385; and US 2011/0200632, the disclosures of which are incorporated herein. [0031] As described above for lipid moieties generally, a palmitic acid adjuvant contains at a minimum at least one palmitic acid moiety, which can be coupled onto an amino acid, an oligopeptide or other molecules. A palmitic acid moiety or a structure containing palmitic acid can be coupled covalently or non-covalently to an antigen to create antigenic compounds with built-in adjuvanting properties. The palmitic acid moiety or a chemical structure containing palmitic acid can be conjugated to a cysteine peptide (Cys) to allow for various structural configurations of the adjuvant, including linear and branched structures. The cysteine residue has been commonly extended by polar residues such as Serine (Ser) and/or lysine (Lys) at the C terminus to create adjuvant compounds with improved solubility. Palmitic acid containing adjuvant compounds could be admixed with an antigen, associated with antigen through non-covalent interactions, or alternatively covalently linked to an antigen, either directly or with the use of a linker/spacer, to generate enhanced immune responses. Most commonly, two
palmitic acid moieties are attached to a glyceryl backbone and a cysteine residue to create dipalmitoyl-S-glyceryl-cysteine (PAM2Cys) or tripalmitoyl-S-glyceryl-cysteine (PAM3Cys), which can also be used in multiple configurations as described above. [0032] In an embodiment, the lipid-based adjuvant is any type of adjuvant comprising a palmitic acid moiety or component. In an embodiment, lipid-based adjuvant is a lipopeptide comprising one or more palmitic acid moieties. The palmitic acid moiety may be modified or manipulated to improve its stability in vitro or in vivo, enhance its binding to receptors (such as for example toll-like receptors as described below) or enhance its biological activity. [0033] In an embodiment, the palmitic acid adjuvant comprises PAM2Cys. In another embodiment, the palmitic acid adjuvant comprises PAM3Cys. [0034] In an embodiment, the palmitic acid adjuvant comprises PAM2-Cys-Ser-(Lys)4 or PAM3-Cys-Ser-(Lys)4. [0035] In some embodiments, the palmitic acid adjuvant is an analog of PAM2-Cys-Ser- (Lys)4 or PAM3-Cys-Ser-(Lys)4 including, without limitation, PAM3Cys-SKKKK (β- irradiated), R-PAM3Cys-SKKKK S-PAM3Cys-SKKKK, PAM3Cys-SKKKK(Biotin-Aca-Aca), PAM3Cys-SKKKK(Fluorescein-Aca-Aca), PAM3Cys-SKKKK, PAM3Cys-SKKKK-FLAG-tag, PAM3Cys-SSNAKIDQLSSDVQT, PAM3Cys-SSNKSTTGSGETTTA, PAM3Cys- SSTKPVSQDTSPKPA, PAM3Cys-SSGSKPSGGPLPDAK, PAM3Cys-SSGNKSAPSSSASSS, PAM3Cys-GSHQMKSEGHANMQL, PAM3Cys-SSSNNDAAGNGAAQT, PAM3Cys-KQNVSSLDEKNSVSV, PAM3Cys-NNSGKDGNTSANSAD, PAM3Cys- NNGGPELKSDEVAKS, PAM3Cys-SQEPAAPAAEATPAG, PAM3Cys- SSSKSSDSSAPKAYG, PAM3Cys-AQEKEAKSELDYDQT, Pam2Cys-SKKKK (mixture of RR and RS stereoisomers), R-Pam2Cys-SKKKK (RR stereoisomer), S-Pam2Cys-SKKKK (RS stereoisomer), PamCys(Pam)-SKKKK, Pam2Cys-SKKKK(Biotin-Aca-Aca)-NH2, Pam2Cys- SKKKK(Fluorescein-Aca-Aca)-NH2, PAM2Cys-SKKKK(Rhodamine-Aca-Aca)-NH2, and PAM2Cys-SKKKK-FLAG-tag. Where appropriate, the palmitic acid adjuvant or analog thereof may used as stereochemically defined compounds or as a mixture of stereoisomers.
[0036] In an embodiment, the lipid-based adjuvant is PAM3-Cys-Ser-(Lys)4:
that act as TLR agonists may also be used as the lipid-based adjuvant disclosed herein, including without limitation the palmitic acid adjuvants and analogs described above and synthetic diacylated lipoprotein FSL-1 available from InvivoGen (San Diego, California, USA) and EMC Microcollections GmbH (Germany). FSL-1 (Pam2CGDPKHPKSF) is a synthetic lipoprotein that represents the N-terminal part of the 44-kDa lipoprotein LP44 of Mycoplasma salivarium. FSL-1 comprises PAM2Cys and has a similar framework structure as macrophage activating lipopeptide-2 (MALP-2), a Mycoplasma fermentans derived lipopeptide. [0038] In an embodiment, the lipid-based adjuvant comprises FSL-1 or MALP-2, or the lipid-based adjuvant is FSL-1 or MALP-2. Where appropriate, FSL-1 or MALP-2 may be used as stereochemically defined compounds or as a mixture of stereoisomers. The FSL-1 or MALP- 2 may be labelled (e.g. biotin, Fluorescein, Rhodamine, etc.). FSL-1 is also available as a FSL-1 Ala-scan collection (EMC Microcollections) comprising nine different FSL-1-Ala compounds. [0039] Further embodiments of lipid-based adjuvants that comprise palmitic acid may include substructures of TLR2 ligands such as monoacylated lipopeptides. Without limitation, these may include, for example, Pam-Dhc-SKKKK, Pam-CSKKKK, Pam-Dhc-GDPKHPKSF or Pam-CGDPKHPKSF (EMC Microcollections). [0040] Therapeutic agent [0041] As used herein, the term “therapeutic agent” is any molecule, substance or compound that is capable of providing a therapeutic activity, response or effect in the treatment or prevention of a disease, disorder or condition, including diagnostic and prophylactic agents. [0042] In relation to the methods disclosed herein providing a dried preparation, a “first therapeutic agent” is any one or more therapeutic agents which are used in the preparation of the non-sized lipid vesicle particle preparation (i.e. incorporated in the methods before the step of sizing the non-sized lipid vesicle preparation). In contrast, a “second therapeutic agent” is any one or more therapeutic agents which are used in the methods herein after preparation of the
sized lipid vesicle particle preparation (i.e. incorporated in the methods after the step of sizing the non-sized lipid vesicle preparation). In embodiments of the methods disclosed herein, lipids may first be dissolved and mixed in an organic solvent. In embodiments where different types of lipid are used, this step will allow a homogenous mixture of the lipids to be formed. In an embodiment, these steps may be carried out in chloroform, chloroform:methanol mixtures, tertiary butanol or cyclohexane. In an embodiment, the lipids are prepared at 10-20mg lipid/mL organic solvent; however, higher or lower concentrations may also be used. In some embodiments, after mixing, the organic solvent is removed (e.g. by evaporation) to yield a lipid film. The lipid film may then be frozen and lyophilized to yield a dry lipid film. The dry lipid film may then be hydrated with an aqueous solution containing therapeutic agents to provide a non-sized lipid vesicle particle preparation. The step of hydration may be performed with shaking and/or mixing (e.g. at 300 RPM for about 1 hour). In yet another embodiment of the methods disclosed herein, an aqueous solution of lipids may be combined with a solution containing one or more solubilized therapeutic agents. In another embodiment, one or more dry therapeutic agents may be added to, and solubilized in, the aqueous solution of lipids or sized lipid vesicle preparation. These embodiments may be performed with shaking and/or mixing (e.g. at 300 RPM for about 1 hour). The above procedures are exemplary methods for providing a non-sized lipid vesicle particle preparation comprising non-sized lipid vesicles and one or more first therapeutic agents. [0043] In some embodiments, therapeutic agents are either solubilized in a solvent (e.g. aqueous or organic) prior to mixing with lipid vesicle particles or therapeutic agents are solubilized upon being mixed with the lipid vesicle particles. In this latter embodiment, therapeutic agents may be added as a dry powder to a solution containing lipid vesicle particles or both the lipid vesicle particles and dry therapeutic agents may be mixed together in a fresh solvent. [0044] In an embodiment, the therapeutic agent is an antigen, a small molecule drug, an antibody or a functional fragment thereof, an antibody mimetic or a functional fragment thereof, an immunomodulatory agent, a polynucleotide encoding a polypeptide, or an interfering polynucleotide. [0045] In some embodiments, the therapeutic agent is an antigen. As used herein, the term “antigen” may be used interchangeably with “immunogen” and may refer to a pathogen, a part of a pathogen, or a molecule that is able to induce an adaptive antibody immune response and/or an adaptive cellular immune response that is specific to said antigen or a portion thereof.
The term “antigen” may refer to a pathogen, a part of a pathogen, or a molecule that is specifically recognized by antibodies and/or immunoglobulin receptors of the adaptive antibody immune response and/or the adaptive cellular immune response. As used herein, the term “peptide antigen” is an antigen as defined above that is a protein or a polypeptide. Adaptive antibody responses are mediated by B cells, which recognize antigen by specific binding of IgM immunoglobulins in their B cell receptors to the antigen. Activated B cells may mature into plasma cells and secrete soluble IgM, IgG, IgA, or IgE antibodies that specifically to bind to the antigen. Cellular immune responses are mediated by CD8+ cytotoxic T cells, which recognize polypeptide antigen by specific binding of T cell receptors (specifically the immunoglobulin superfamily TCR-alpha/TCR-beta or TCR-gamma/TCR-delta heterodimers) to peptides from the antigen that are displayed on MHC class I molecules on the surface of target cells. Antibody and cellular adaptive immune responses are coordinated by CD4+ T helper cells, which recognize polypeptide antigen by specific binding of their T cell receptors (specifically the immunoglobulin superfamily TCR-alpha/TCR-beta or TCR-gamma/TCR-delta heterodimers) to peptides from the antigen that are displayed on MHC class I/II molecules on the surface of APC. The immunoglobulin superfamily proteins in B cell receptors and antibodies (IgM, IgD, IgG, IgA, IgE), and in T cell receptors (TCR-alpha/TCR-beta, TCR-gamma/TCR-delta) that specifically bind to antigen are created randomly by processes of genetic recombination during B cell and T cell development. Each B cell and T cell expresses only one species of randomized immunoglobulin superfamily receptor with a specific binding recognition, and B cells and T cells that bind an antigen are clonally selected for expansion in the body during an immune response. Further, the immunoglobulins in B cell receptors and antibodies may further diversify by somatic mutation and clonal selection for variants with high antigen binding. By these processes, antibodies, B cell receptors, and T cell receptors are immunoglobulin superfamily molecules that enable the immune system to adapt to an antigen, thus forming an adaptive immune response. This adaptive immune response contrasts with innate immune responses that are mediated by conserved immune receptors, such as TLR or other pattern recognition receptors, that are unchanging and have fixed specificity for binding to specific pathogen- associated molecular patterns. Thus, as used herein, the term “antigen” or “peptide antigen” may refer to a molecule that is specifically bound by antibodies, B cell receptors, and/or T cell receptors of the adaptive immune system during an active adaptive immune response. The specific portion of an antigen that is bound by an antibody, a B cell receptor, and/or a T cell receptor is called an “epitope”. Antibodies and B cell receptors may bind to an epitope on various types of molecules including polypeptides, polysaccharides, glycoproteins, and lipoproteins. T cell receptors bind to peptides that are derived from antigen, wherein the peptides
are displayed on MHC class I/II molecules. Thus, the term “antigen” may refer to a molecule that comprises a B cell epitope and/or a T cell epitope. [0046] In some embodiments, the antigen may include a polypeptide, a polysaccharide, a glycoprotein, a lipoprotein, a microorganism or a part thereof, such as a live, attenuated, inactivated or killed bacterium, virus or protozoan, or part thereof, an allergen, or an antigen derived from a cancer cell (such as a conserved cancer antigen or a neoantigen). As used herein, the term “derived from” encompasses, without limitation: an antigen that is isolated or obtained directly from an originating source; a synthetic or recombinantly generated antigen that is identical or substantially related to an antigen from an originating source; or an antigen which is made from an antigen of an originating source or a fragment thereof. When it is stated that an antigen is “from” a source, the term “from” may be equated with “derived from”. The term “substantially related”, as this context, means that the antigen may have been modified by chemical, physical or other means (e.g. sequence modification), but that the resultant product remains capable of generating an immune response to the original antigen or to the disease or disorder associated with the original antigen. As used herein, the term “antigen” also includes a polynucleotide that encodes a polypeptide that functions as an antigen. Nucleic acid-based vaccination strategies are known, wherein a vaccine composition that contains a polynucleotide is administered to a subject. The antigenic polypeptide encoded by the polynucleotide is expressed in the subject, such that the antigenic polypeptide is ultimately present in the subject. [0047] Viruses, or parts thereof, from which a peptide antigen may be derived include for example, and without limitation, Cowpoxvirus, Vaccinia virus, Pseudocowpox virus, herpes virus, Human herpesvirus 1, Human herpesvirus 2, Cytomegalovirus, Human adenovirus A-F, Polyomavirus, human papillomavirus (HPV), Parvovirus, Hepatitis A virus, Hepatitis B virus, Hepatitis C virus, human immunodeficiency virus (HIV), Seneca Valley virus (SVV), Orthoreovirus, Rotavirus, Ebola virus, parainfluenza virus, influenza virus (e.g. H5N1 influenza virus, influenza A virus, influenza B virus, influenza C virus), Measles virus, Mumps virus, Rubella virus, Pneumovirus, respiratory syncytial virus, respiratory syncytial virus (RSV), Rabies virus, California encephalitis virus, Japanese encephalitis virus, Hantaan virus, Lymphocytic choriomeningitis virus, Coronavirus (e.g. Sars-Cov-2), Enterovirus, Rhinovirus, Poliovirus, Norovirus, Flavivirus, Dengue virus, West Nile virus, Yellow fever virus and varicella. [0048] In an embodiment, the peptide antigen is derived from HPV. In an embodiment, the HPV peptide antigen is one that is associated with HPV-related cervical cancer or
HPV-related head and neck cancer. In an embodiment, the peptide antigen is a peptide comprising the sequence RAHYNIVTF (HPV16E7 (H-2Db) peptide 49-57; R9F). In an embodiment, the peptide antigen is a peptide comprising the sequence YMLNLGPET (HPV Y9T peptide). [0049] In an embodiment, the peptide antigen is derived from HIV. In an embodiment, the HIV peptide antigen may be derived from the V3 loop of HIV-1 gp120. In an embodiment, the HIV peptide antigen may be RGP10 (RGPGRAFVTI). In another embodiment, the peptide antigen may be AMQ9 (AMQMLKETI). AMQ9 peptide is the immunodominant MHC class I epitope of gag for mice of the H-2Kd haplotype. [0050] In an embodiment, the peptide antigen is derived from RSV. The RSV virion, a member of the genus Paramyxoviridae, is composed of a single strand of negative-sense RNA with 15,222 nucleotides. The nucleotides encode three transmembrane surface proteins (F, G and small hydrophobic protein or SH), two matrix proteins (M and M2), three nucleocapsid proteins (N, P and L), and two non-structural proteins (NS1 and NS2). In an embodiment, the peptide antigen may be derived from any one or more of the RSV proteins. In a particular embodiment, the peptide antigen may be derived from the SH protein of RSV or any other paramyxovirus, or a fragment thereof. The RSV peptide antigen may be any one or more of the RSV peptides described or disclosed in WO 2012/065997, incorporated herein by reference. [0051] Bacteria, or parts thereof, from which a peptide antigen may be derived include for example, and without limitation, Anthrax (Bacillus anthracis), Brucella, Bordetella pertussis, Candida, Chlamydia pneumoniae, Chlamydia psittaci, Cholera, Clostridium botulinum, Coccidioides immitis, Cryptococcus, Diphtheria, Escherichia coli O157: H7, Enterohemorrhagic Escherichia coli, Enterotoxigenic Escherichia coli, Haemophilus influenzae, Helicobacter pylori, Legionella, Leptospira, Listeria, Meningococcus, Mycoplasma pneumoniae, Mycobacterium, Pertussis, Pneumonia, Salmonella, Shigella, Staphylococcus, Streptococcus pneumoniae and Yersinia enterocolitica. [0052] In an embodiment, the peptide antigen is derived from a Bacillus anthracis. Without limitation, the peptide antigen may for example be derived from anthrax recombinant protective antigen (rPA) (List Biological Laboratories, Inc.; Campbell, CA) or anthrax mutant recombinant protective antigen (mrPA). rPA has an approximate molecular weight of 83,000 daltons (Da) and corresponds a cell binding component of the three-protein exotoxin produced by Bacillus anthracis. The protective antigen mediates the entry of anthrax lethal factor and edema factor into the target cell. In some embodiments the antigen may be derived from the
sequence found under GenBank Accession number P13423, or any suitable sequence variant thereof. [0053] Protozoa, or parts thereof, from which a peptide antigen may be derived include for example, and without limitation, the genus Plasmodium (Plasmodium falciparum, Plasmodium malariae, Plasmodium vivax, Plasmodium ovale or Plasmodium knowlesi), which causes malaria. [0054] In an embodiment, the peptide antigen is derived from a Plasmodium species. For example, and without limitation, the peptide antigen may be derived from the circumsporozoite protein (CSP), which is a secreted protein of the sporozoite stage of the malaria parasite (Plasmodium sp.). The amino-acid sequence of CSP consists of an immunodominant central repeat region flanked by conserved motifs at the N- and C-termini that are implicated in protein processing as the parasite travels from the mosquito to the mammalian vector. The structure and function of CSP is highly conserved across the various strains of malaria that infect humans, non-human primates and rodents. In an embodiment, the peptide antigen derived from CSP is a malaria virus-like particle (VLP) antigen which comprises circumsporozoite T and B cell epitopes displayed on the woodchuck hepatitis virus core antigen. [0055] In another embodiment, the peptide antigen may be derived from a cancer or tumor-associated protein, such as for example, a membrane surface-bound cancer antigen. In an embodiment, the cancer may be one that is caused by a pathogen, such as a virus. Viruses linked to the development of cancer are known to the skilled person and include, but are not limited to, human papillomaviruses (HPV), John Cunningham virus (JCV), Human herpes virus 8, Epstein Barr Virus (EBV), Merkel cell polyomavirus, Hepatitis C Virus and Human T cell leukaemia virus-1. Thus, in an embodiment, the peptide antigen may be derived from a virus that is linked to the development of cancer. In an embodiment, the peptide antigen is a cancer-associated antigen. Many cancer or tumor-associated proteins are known in the art such as for example, and without limitation, those described in WO 2016/176761, incorporated by reference herein. [0056] In an embodiment, the peptide antigen is one or more survivin antigens. Survivin, also called baculoviral inhibitor of apoptosis repeat-containing 5 (BIRC5), is a protein involved in the negative regulation of apoptosis. In an embodiment, the peptide antigen is any peptide, polypeptide or variant thereof derived from a survivin protein, or a fragment thereof. In an embodiment, the survivin peptide antigen may comprise the full length survivin polypeptide. Alternatively, the survivin peptide antigen may be a survivin peptide comprising a fragment of any length of the survivin protein. Exemplary embodiments include a survivin peptide that
comprises at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 amino acid residues. In specific embodiments, the survivin peptide consists of a heptapeptide, an octapeptide, a nonapeptide, a decapeptide or an undecapeptide, consisting of 7, 8, 9, 10, 11 consecutive amino acid residues of the survivin protein, respectively. Particular embodiments of the survivin antigen include survivin peptides of about 9 or 10 amino acids. Survivin peptide antigens also encompass variants and functional equivalents of natural survivin peptides. Variants or functional equivalents of a survivin peptide encompass peptides that exhibit amino acid sequences with differences as compared to the specific sequence of the survivin protein, such as one or more amino acid substitutions, deletions or additions, or any combination thereof. The difference may be measured as a reduction in identity as between the survivin protein sequence and the survivin peptide variant or survivin peptide functional equivalent. In an embodiment, a pharmaceutical composition of the present invention may include any one or more of the survivin peptides, survivin peptide variants or survivin peptide functional equivalents disclosed in WO 2004/067023; WO 2006/081826 or WO 2016/176761, each of which is incorporated by reference herein. In an embodiment, the survivin peptide antigen may be any one or more of FEELTLGEF, FTELTLGEF, LTLGEFLKL, LMLGEFLKL, RISTFKNWPF, RISTFKNWPK, STFKNWPFL, and/or LPPAWQPFL. [0057] In an embodiment, the peptide antigen is a neoantigen. As used herein, the term “neoantigen” refers to a class of tumor antigens which arise from tumor-specific mutations in an expressed protein. The neoantigen can be derived from any cancer, tumor or cell thereof. In the context of neoantigens, the term “derived from” as used herein encompasses, without limitation: a neoantigen that is isolated or obtained directly from an originating source (e.g. a subject); a synthetic or recombinantly generated neoantigen that is identical in sequence to a neoantigen from an originating source; or a neoantigen which is made from a neoantigen of an originating source or a fragment thereof. The mutations in the expressed protein that create the neoantigen may be patient-specific. By “patient-specific”, it is meant that the mutation(s) are unique to an individual subject. However, it is possible that more than one subject will share the same mutation(s). Thus, a “patient-specific” mutation may be shared by a small or large sub-population of subjects. The neoantigen may comprise one or more neoepitopes. As used herein, the term “epitope” refers to a peptide sequence which can be recognized by the immune system, specifically by antibodies, B cells or T cells. A “neoepitope” is an epitope of a neoantigen which comprises a tumor-specific mutation as compared to the native amino acid sequence. Generally, neoepitopes may be identified by screening neoantigens for anchor residues that have the potential to bind patient HLA. The neoepitopes are normally ranked using
algorithms, such as NetMHC, that can predict peptide binding to HLA. A "T-cell neoepitope" is to be understood as meaning a mutated peptide sequence which can be bound by the MHC molecules of class I or II in the form of a peptide-presenting MHC molecule or MHC complex. The T-cell neoepitope should typically be one that is amenable to recognition by T cell receptors so that a cell-mediated immune response can occur. A "B-cell neoepitope" is to be understood as meaning a mutated peptide sequence which can be recognized by B cells and/or by antibodies. In some embodiments, at least one of the neoepitopes of the neoantigen is a patient-specific neoepitope. As used herein, by “patient-specific neoepitope”, it is meant that the mutation(s) in the neoepitope are unique to an individual subject. However, it is possible that more than one subject will share the same mutation(s). Thus, a “patient-specific neoepitope” may be shared by a small or large sub-population of subjects. In an embodiment, the neoantigen may be derived from a mutated gene or protein that has previously been associated with cancer phenotypes, such as for example tumor suppressor genes (e.g. p53); DNA repair pathway proteins (e.g. BRCA2) and oncogenes. In some embodiments, the neoantigen may comprise or consist of the neoantigens disclosed in Castle et al. (2012) Exploiting the Mutanome for Tumor Vaccination. Cancer Res, 72(5): 1081-1091, incorporated by reference herein. In an embodiment, the neoantigen may be one or more of the Mut1-50 neoantigens disclosed in Table 1 of Castle 2012, or a neoantigen of the same or related protein (e.g. a human homologue). In an embodiment, the neoantigen may be one or more of Mut25 (STANYNTSHLNNDVWQIFENPVDWKEK), Mut30 (PSKPSFQEFVDWENVSPELNSTDQPFL) and Mut44 (EFKHIKAFDRTFANNPGPMVVFATPGM), or a neoantigen of the same or related protein (e.g. a human homologue). [0058] Many peptide antigens are known in the art and may be suitable for use as therapeutic agents of the present invention. In an embodiment, the antigen may be a peptide derived from the ectodomain of the small hydrophobic protein of respiratory syncytial virus (RSV) as disclosed in WO2012/065997, incorporated herein by reference. For example, the RSV antigen may be a peptide with the sequence NKLCEYNVFHNKTFELPRARVNT, NKLSEHKTFCNNTLELGQMHQINT, or NKLCDFNDHHTNSLDIRTRLRNDTQLITRAHEGSINQSSN, or a portion or variant thereof, In an embodiment, the antigen may be a peptide derived from the survivin protein as disclosed in WO2004/067023 and WO2006/081826, incorporated herein by reference. For example, the survivin antigen may be one or more peptides of the sequence FEELTLGEF, FTELTLGEF, LTLGEFLKL, LMLGEFLKL, RISTFKNWPF, RISTFKNWPK, STFKNWPFL, or LPPAWQPFL, or a portion or a variant thereof. In an embodiment, the antigen may be a fusion
peptide (FP) antigen comprising the sequence RAHYNIVTF (HPV16E7 (H-2Db) peptide 49-57) fused to the universal T-helper epitope is PADRE (pan-DR epitope) comprising the peptide sequence AKXVAAWTLKAAA, wherein X may be phenylalanine or cyclohexylalanine. [0059] In a particular embodiment, the therapeutic agent is one or more peptide antigens as described herein. In an embodiment, the peptide antigen is a synthetically produced polypeptide. The peptide antigen may be a polypeptide of any length. In an embodiment, the peptide antigen may be 5 to 120 amino acids in length, 5 to 100 amino acids in length, 5 to 75 amino acids in length, 5 to 50 amino acids in length, or 5 to 30 amino acids in length. In an embodiment, the peptide antigen may be 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49 or 50 amino acids in length. In an embodiment, the peptide antigen is 20 to 30 amino acids in length. In an embodiment, the peptide antigen is 27 amino acids in length. In an embodiment, the peptide antigen is 8 to 40 amino acids in length. In an embodiment, the peptide antigen is 9 or 10 amino acids in length. [0060] In some embodiments, the therapeutic agent is a small molecule drug. The term “small molecule drug” refers an organic or inorganic compound that may be used to treat, cure, prevent or diagnose a disease, disorder or condition. As used herein, the term "small molecule" refers to a low molecular weight compound which may be synthetically produced or obtained from natural sources and has a molecular weight of less than 2000 Daltons (Da), less than 1500 Da, less than 1000 Da, less than 900 Da, less than 800 Da, less than 700 Da, less than 600 Da or less than 500 Da. A small molecule drug is typically a chemically manufactured active substance or compound (i.e. it is not produced by a biological process). Generally, these compounds are synthesized by chemical reactions between different organic and/or inorganic compounds. As used herein, the term “small molecule drug” does not encompass larger structures, such as polynucleotides, proteins, and polysaccharides, which are made by a biological process. The term “small molecule” may refer to compounds or molecules that selectively bind specific biological macromolecules and act as an effector, altering the activity or function of the target. Thus, a small molecule drug is a substance or compound that regulates a biological process in the body of a subject, and more particularly within a cell. A small molecule drug may exert its activity in the form in which it is administered, or the small molecule drug may be a prodrug. In this regard, the term “small molecule drug”, as used herein, encompasses both the active form and the prodrug. The term “prodrug” refers to a compound or substance that, under physiological conditions, is converted into the therapeutically active agent. A prodrug is a compound or substance that, after administration, is metabolized in the body of a subject into the
pharmaceutically active form (e.g. by enzymatic activity in the body of the subject). A common method for making a prodrug is to include selected moieties that are hydrolyzed under physiological conditions to reveal the pharmaceutically active form. Many small molecule drugs are known in the art and are used as active ingredients in medicaments. The skilled person is aware of numerous small molecule drugs such as those disclosed in the online DrugBank database. A small molecule drug may include a cytotoxic agent, an anti-cancer agent, an anti- tumor agent, a chemotherapeutic agent, an anti-neoplastic agent, an antiviral agent, an antibacterial agent, an anti-inflammatory agent, an immunomodulatory agent (e.g. a cytokine or a chemokine), an immune response checkpoint agent, a biological response modifier, a prodrug, a ligand, an analgesic, a radiopharmaceutical, a radioisotope or a dye for visual detection. A cytotoxic agent may be an agent that kills a target cell by necrosis or apoptosis. A cytotoxic agent may include epacadostat, cyclophosphamide, rapamycin, ifosfamide, afosfamide, melphalan, bendamustine, uramustine, palifosfamide, chlorambucil, busulfan, 4-hydroxycyclophosphamide, bis-chloroethylnitrosourea (BCNU), mitomycin C, yondelis, procarbazine, dacarbazine, temozolomide, cisplatin, carboplatin, oxaliplatin, acyclovir, gemcitabine, 5-fluorouracil, cytosine arabinoside, ganciclovir, camptothecin, topotecan, irinotecan, doxorubicin, daunorubicin, epirubicin, idarubicin, etoposide, teniposide, mitoxantrone, valproic acid, vorinostat, methotrexate, tacrolimus, or pixantrone. A small molecule drug may include an checkpoint agent. As used herein, a “checkpoint agent” refers to any compound or molecule that totally or partially modulates (e.g. activates or inhibits) the activity or function of one or more checkpoint molecules (e.g. proteins). Checkpoint molecules regulate various cellular processes. A small molecule drug may include a checkpoint agent that is an inhibitor of Programmed Death-Ligand 1 (PD-L1, also known as B7-H1, CD274), Programmed Death 1 (PD-1, CD279), CTLA-4 (CD154), PD-L2 (B7-DC, CD273), LAG3 (CD223), TIM3 (HAVCR2, CD366), 41BB (CD137), 2B4, A2aR, B7H1, B7H3, B7H4, B- and T-lymphocyte attenuator (BTLA), CD2, CD27, CD28, CD30, CD33, CD40, CD70, CD80, CD86, CD160, CD226, CD276, DR3, GAL9, GITR, HVEM, IDO1, IDO2, ICOS (inducible T cell costimulator), Killer inhibitory receptor (KIR), LAG-3, LAIR1, LIGHT, MARCO (macrophage receptor with collageneous structure), phosphatidylserine (PS), OX-40, Siglec-5, Siglec-7, Siglec-9, Siglec-11, SLAM, TIGIT, TIM3, TNF-α, VISTA, VTCN1, or any combination thereof. A small molecule drug may include a cell-penetrating peptide, a peptide transduction domain, or a dendritic cell peptide, used as molecular shuttles that can transport other molecules or ions from one location to another.
[0061] In some embodiments, the therapeutic agent is an antibody, a functional equivalent of an antibody or a functional fragment of an antibody. Broadly, an “antibody” refers to a polypeptide or protein that consists of or comprises antibody domains, which are understood as constant and/or variable domains of the heavy and/or light chains of immunoglobulins, with or without a linker sequence. Antibody domains may be of native structure or modified by mutagenesis or derivatization, e.g. to modify binding specificity or any other property. An antibody may comprise a complete (i.e. full-length) immunoglobulin molecule, including e.g. polyclonal, monoclonal, chimeric, humanized and/or human versions having full length heavy and/or light chains. The term “antibody” encompasses any and all isotypes and subclasses, including without limitation the major classes of IgA, IgD, IgE, IgG and IgM, and the subclasses IgG1, IgG2, IgG3, IgG4, IgA1 and IgA2. An antibody may be one that is naturally occurring or one that is prepared by any means available to the skilled person, such as for example by using immunoglobulin gene fragment recombinatorial processes. An antibody may be a monoclonal antibody, a polyclonal antibody, a chimeric antibody, a humanized antibody, a human antibody or a fully human antibody. A “chimeric antibody” as used herein refers to a recombinant protein that contains the variable domains (including the complementarity determining regions (CDRs)) of an antibody derived from one species, such for example a rodent, while the constant domains of the antibody are derived from a different species, such as a human. For veterinary applications, the constant domains of the chimeric antibody may be derived from that of an animal, such as for example a cat or dog. A “humanized antibody” as used herein refers to a recombinant protein in which the CDRs from an antibody from one species; e.g., a rodent, are transferred from the heavy and light variable chains of the rodent antibody into human heavy and light variable domains, including human framework region (FR) sequences. The constant domains of the humanized antibody are likewise derived from a human antibody. Without limitation, a “human antibody” as used herein refers to an antibody obtained from transgenic animals (e.g. mice) that have been genetically engineered to produce specific human antibodies in response to antigenic challenge. In this technique, elements of the human heavy and light chain loci are introduced into strains of mice derived from embryonic stem cell lines that contain targeted disruptions of the endogenous heavy chain and light chain loci. The transgenic animal can synthesize human antibodies specific for human antigens, and the animal can be used to produce human antibody-secreting hybridomas. A fully human antibody also can be constructed by genetic or chromosomal transfection methods, as well as phage display technology, all of which are known in the art. As used herein, the term “functional fragment”, with respect to an antibody, refers to an antigen-binding portion of an antibody. In this context, by “functional” it is meant that the fragment maintains its ability to bind to the target antigen.
Functional fragments of antibodies include a portion of an antibody such as a F(ab')2, a F(ab)2, a Fab', a Fab, a Fab2, a Fab3, and single domain antibody. Regardless of structure, a functional fragment of an antibody binds with the same antigen that is recognized by the intact antibody. The term “functional fragment”, in relation to antibodies, also includes isolated fragments consisting of the variable regions, such as the “Fv” fragments consisting of the variable regions of the heavy and light chains and recombinant single chain polypeptide molecules in which light and heavy chain variable regions are connected by a peptide linker (“scFv proteins”). Antibody fragments can be incorporated into single domain antibodies (e.g. nanobodies), single-chain antibodies, maxibodies, evibodies, minibodies, intrabodies, diabodies, triabodies, tetrabodies, vNAR, bis-scFv and other like structures. Another form of a functional fragment is a peptide comprising one or more CDRs of an antibody or one or more portions of the CDRs, provided the resultant peptide retains the ability to bind the target antigen. Antibodies for therapeutic use are known in the art such as, for example, anti-CTLA4 antibodies (ipilimumab, tremelimumab, BN- 13, UC10-4F10-11, 9D9 or 9H10), anti-PD1 antibodies (pembrolizumab, nivolumab, pidilizumab, AMP-224, RMP1-4 or J43), and anti-PDL1 antibodies (tezolizumab, avelumab, BMS-936559 or durvalumab). [0062] In some embodiments, the therapeutic agent is an antibody mimetic, a functional equivalent of an antibody mimetic, or a functional fragment of an antibody mimetic. As used herein, the term “antibody mimetic” refers to compounds which, like antibodies, can specifically and/or selectively bind antigens or other targets, but which are not structurally related to antibodies. Antibody mimetics are usually artificial peptides or proteins and are typically smaller than antibodies, with a molar mass of about 3-20 kDa (whereas antibodies are generally about 150 kDa). Non-limiting examples of antibody mimetics include peptide aptamers, affimers, affilins, affibodies, affitins, alphabodies, anticalins, avimers, DARPins, fynomers, Kunitz domain peptides, nanoCLAMPs, monobodies, affinity reagents and scaffold proteins. As used herein, the term “functional fragment”, with respect to an antibody mimetic, refers any portion or fragment of an antibody mimetic that maintains the ability to bind to its target molecule. The functional fragment of an antibody mimetic may be, for example, a portion of any of the antibody mimetics as described herein. As used herein, a “functional equivalent” in the context of an antibody mimetic refers to a polypeptide or other compound or molecule having similar binding characteristics to an antibody mimetic, but not necessarily being a recognizable “fragment” of an antibody mimetic. [0063] In some embodiments, the therapeutic agent is an immunomodulatory agent. As used herein, an “immunomodulatory agent” is a molecule or compound that modulates the
activity and/or effectiveness of an immune response. “Modulate”, as used herein, means to enhance (upregulate), suppress (downregulate), direct, redirect or reprogram an immune response. The term “modulate” is not intended to mean activate or induce. By this, it is meant that the immunomodulatory agent modulates (enhances, reduces or directs) an immune response that is activated, initiated or induced by a particular antigen, but the immunomodulatory agent is not itself the antigen against which the immune response is directed, nor is the immunomodulatory agent derived from that antigen. An immunomodulatory agent that enhances the immune response may be selected from cytokines (e.g. certain interleukins and interferons), stem cell growth factors, lymphotoxins, co-stimulatory molecules, hematopoietic factors, colony stimulating factors, erythropoietins, thrombopoietins, and the like, and synthetic analogs of these molecules. An immunomodulatory agent may include: lymphotoxins, such as tumor necrosis factor (TNF); hematopoietic factors, such as interleukin (IL); colony stimulating factor, such as granulocyte-colony stimulating factor (G-CSF) or granulocyte macrophage-colony stimulating factor (GM-CSF); interferon, such as interferons-alpha, -beta or –lamda; and stem cell growth factor, such as that designated "SI factor". Included among the cytokines are hormones, such as human growth hormone, N-methionyl human growth hormone, and bovine growth hormone; parathyroid hormone; thyroxine; insulin; proinsulin; relaxin; prorelaxin; glycoprotein hormones, such as follicle stimulating hormone (FSH), thyroid stimulating hormone (TSH), and luteinizing hormone (LH); hepatic growth factor; prostaglandin, fibroblast growth factor; prolactin; placental lactogen, OB protein; tumor necrosis factor-alpha and -beta; mullerian-inhibiting substance; mouse gonadotropin-associated peptide; inhibin; activin; vascular endothelial growth factor; integrin; thrombopoietin (TPO); nerve growth factors, such as NGF-beta; platelet-growth factor; transforming growth factors (TGFs), such as TGF-alpha and TGFP; insulin-like growth factor-I and -II; erythropoietin (EPO); osteoinductive factors; interferons, such as interferon-alpha, -beta, and -gamma; colony stimulating factors (CSFs), such as macrophage-CSF (M-CSF); interleukins (ILs), such as IL-1, IL-lalpha, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL- 9, IL-10, IL-11, IL-12; IL-13, IL-14, IL-15, IL-16, IL-17, IL-18, IL-21, IL-25, LIF, kit-ligand or FLT-3, angiostatin, thrombospondin, endostatin and tumor necrosis factor. An immunomodulatory agent may be an immune costimulatory molecule agonist. Immune costimulatory molecules are signaling proteins that play a role in regulating immune response. Some immune costimulatory molecules are receptors located on the surface of a cell that respond to extracellular signaling. When activated, immune costimulatory molecules produce a pro-inflammatory response that can include suppression of regulatory T cells and activation of cytotoxic or killer T cells. Exemplary immune costimulatory molecules include CD27, CD28, CD40, CD122, CD137, CD137/4-1BB, ICOS, IL-10, OX40 TGF-beta, TOR
receptor, and glucocorticoid-induced TNFR-related protein GITR. An immune costimulatory molecule agonist may therefore be any compound, molecule or substance that is an agonist of a costimulatory immune molecule as described herein. An immunomodulatory agent may be an immunosuppressive agent. By “immunosuppressive agent”, it is meant a molecule or compound that reduces (downregulates) the activity and/or efficacy of the immune response, or directs, redirects or reprograms the immune response in a manner that alleviates an undesired result (e.g. an autoimmune response or allergy). There are many different types of immunosuppressive agent, including calcineurin inhibitors, interleukin inhibitors, selective immunosuppressants and THF-alpha inhibitors. An immunomodulatory agent may be an immunosuppressant selected from 5-fluorouracil, 6-thioguanine, adalimumab, anakinra, Atgam, abatacept, alefacept, azathioprine, basiliximab, belatacept, belimumab, benralizumab, brodalumab, canakinumab, certolizumab, chlorambucil, cyclosporine, daclizumab, dimethyl fumerate, dupilumab, eculizumab, efalizumab, ethanercept, everolimus, fingolimod, golimumab, guselkumab, imiquimod, infliximab, ixekizumab, leflunomide, lenlidomide, mechlorethamine, mepolizumab, methotrexate, muromonab-cd3, mycophenolate mofetil, mycophenolic acid, natallizumab, omalizumab, pomalidomide, pimecrolimus, reslizumab, rilonacept, sarilumab, secukinumab, siltuximab, sirolimus, tacrolimus, teriflunomide, thalidomide, Thymoglobulin, tocilizumab, ustekinumab, and vedolizumab. An immunomodulatory agent may be any molecule or compound that is an immunosuppressive cytotoxic drug. An immunosuppressive cytotoxic drug may include a glucocorticoid, a cytostatic (e.g. alkylating agents, antimetabolites), an antibody, a drug acting on immunophilins, an interferon, an opioid, or a TNF binding protein. Immunosuppressive cytotoxic drugs include nitrogen mustards (e.g. cyclophosphamide), nitrosoureas, platinum compounds, folic acid analogs (e.g. methotrexate), purine analogs (e.g. azathioprine and mercaptopurine), pyrimidine analogs (e.g. fluorouracil), protein synthesis inhibitors, cytotoxic antibiotics (e.g. dactinomycin, anthracyclines, mitomycin C, bleomycin and mithramycin), cyclosporine, tacrolimus, sirolimus/rapamycin, everolimus, prednisone, dexamethasone, hydrocortisone, mechlorethamine, clorambucil, mycopholic acid, fingolimod, myriocin, infliximab, etanercept, or adalimumab. An immunomodulatory agent may be an anti- inflammatory agent. An anti-inflammatory agent may be a non-steroidal anti-inflammatory agent (such as a Cox-1 and/or Cox-2 inhibitor), a non-steroidal anti-inflammatory agent, aspirin, salsalate, diflunisal, ibuprofen, fenoprofen, flubiprofen, fenamate, ketoprofen, nabumetone, piroxicam, naproxen, diclofenac, indomethacin, sulindac, tolmetin, etodolac, ketorolac, oxaprozin, a corticosteroid, or celecoxib. An immunomodulatory agent may be an anti-rheumatic agent such as prednisone, dexamethasone, chloroquine, hydroxychloroquine, methotrexate, sulfasalazine, cyclosporine, azathioprine, cyclophosphamide, azathioprine, sulfasalazine,
penicillamine, aurothioglucose, gold sodium thiomalate, auranofin, methotrexate, mechlorethamine, cyclophosphamide, chlorambucil, or azathioprine. [0064] In some embodiments, the therapeutic agent is a polynucleotide encoding a polypeptide, or an interfering polynucleotide (such as miRNA, siRNA shRNA, DsiRNA, or a polynucleotide encoding any thereof). A therapeutic agent may be a polynucleotide that contains sequences that correspond largely to the sense or antisense sequence of specific genes or their products, and hence have a direct effect on the expression of these genes and/or their products. For example, the use of polynucleotides that contain gene coding sequences affects the transcription and/or translation of the genes of interest in cells that uptake such polynucleotides. Similarly, the use of RNA interference polynucleotides (miRNA, siRNA, shRNA) affects the expression of specific genes of interest by directly affecting the levels of mRNA in cells that uptake such nucleotides. This differs significantly from other polynucleotide-based molecules such as CpG, and DNA- or RNA- based poly I:C adjuvants, which do not act through the presence of gene specific sequences. Thus, an active agent may be a polynucleotide that is not expressed as a protein in a cell, but rather encodes, for example, an antisense RNA, an interfering RNA, a catalytic RNA, or a ribozyme. RNA interference (RNAi) is a sequence specific post-transcriptional gene silencing mechanism, which is triggered by double-stranded RNA such as small (or short) interference RNA (siRNA), short hairpin RNA (shRNA), and single stranded intracellular RNA such as microRNA (miRNA), all of which can cause degradation of homologous mRNAs in a cell. Interfering RNA may be a naturally occurring or synthetic RNA chain of varying length. Interfering RNA can be duplexes, usually but not always limited to, 20 to 25-nt long that have 19 base pair central double stranded domain with terminal 2-base 3’ overhangs. Interfering RNA can be further modified chemically to enhance its in vivo efficacy, induce nuclease-resistance to prevent degradation and enhance stability. In this regard, the anti-sense strand may have either a free 5’-OH or 5’-phosphate terminus, the latter results in natural Dicer processing and represents the active form of the molecule. Interfering RNA may have phosphorothioate or boranohosphate modification of the internucleoside linkage to improve nuclease stability and prolong life of the duplex when exposed to serum or other nuclease sources. Interfering RNA may have modifications at 2’position, for example, 2’-O-methyl RNA residue incorporation to retain full potency compared with unmodified RNA, retaining stability in serum and significantly reducing the risk of potential IFN responses in the cell. Interfering RNA may also have 2’-fluoro modification, which is usually incorporated selectively at pyrimidine bases, to improve stability and potency.
[0065] In some embodiments, the therapeutic agent is a T-helper peptide. As used herein, the term “T-helper peptide” refers to a peptide that is an epitope strongly recognized by T cells when displayed on MHC class I/II molecules. T-helper peptides are recognized by helper T cells (CD4+ T cells), which play an important role in establishing and maximizing the capabilities of the immune system, and are involved in activating and directing other immune cells, such as for example cytotoxic T cells and B cells. Accordingly, T-helper peptides are capable of enhancing or stimulating an immune response to an antigen. Immunodominant T-helper peptides or universal T-helper peptides are known peptides that are broadly reactive in animal and human populations with widely divergent MHC types. T-helper peptides may be comprised in an antigen, or provided with an antigen to boost the immune response to the antigen. In some embodiments, a T helper peptide may include an immunodominant or universal T helper peptide as known in the art such as, for example but not limited to the modified Tetanus toxin peptide A16L (830-844, AQYIKANSKFIGITEL, with an alanine residue added to its amino terminus to enhance stability); PADRE (pan-DR epitope) comprising the peptide sequence AKXVAAWTLKAAA, wherein X may be phenylalanine or cyclohexylalanine; or Tetanus toxin peptide F21E (947-967, FNNFTVSFWLRVPKVSASHLE). [0066] Hydrophobic carrier [0067] The pharmaceutical compositions of the present comprise a hydrophobic carrier. The hydrophobic carrier may be an essentially pure hydrophobic substance or a mixture of hydrophobic substances. Hydrophobic substances that are useful in the compositions described herein are those that are pharmaceutically acceptable. The hydrophobic carrier is typically a liquid but certain hydrophobic carriers that are not liquids at standard room temperature (about 18-25oC) may be liquefied, for example by warming, and may also be useful. [0068] In some embodiments, the hydrophobic carrier is an oil or a mixture of oils. In some embodiments, the hydrophobic carrier is a mineral oil (such as DrakeolTM 6VR or IFA), a vegetable oil (such as soybean oil), a nut oil (such as peanut oil), or a mixture of any thereof. In some embodiments, the hydrophobic carrier is Incomplete Freund’s Adjuvant (IFA), a mineral oil-based hydrophobic carrier. In some embodiments, the hydrophobic carrier is mannide monooleate in mineral oil, such as commercially available MontanideTM ISA 51 (SEPPIC, France). MontanideTM ISA 51 is a mixture of highly purified mineral oil (DrakeolTM 6VR) and mannide monooleate. In another embodiment, the hydrophobic carrier is mannide oleate in non- mineral oil, such as commercially available MontanideTM ISA 720 (SEPPIC, France). In another embodiment, the hydrophobic carrier is MS80 oil which is a mixture of mineral oil (Sigma
Aldrich) and a sorbitan monooleate (such as SpanTM 80), the components of which can be purchased separately and mixed prior to use. [0069] In the compositions of the present invention, the amount of hydrophobic carrier used will depend on the desired volume of the final composition and/or the desired concentration of the components suspended and/or solubilized in the hydrophobic carrier. [0070] In some embodiments, the compositions of the present invention are water-free or substantially free of water, meaning that the compositions are not emulsions. By “water-free” it is meant that the compositions contain no water at all. In another embodiment, the compositions may be substantially free of water. The term “substantially free of water” is intended to encompass embodiments where the hydrophobic carrier may still contain small quantities of water, provided that the water is present in the non-continuous phase of the carrier. For example, individual components of the composition may have small quantities of bound water that may not be completely removed by processes such as lyophilization or evaporation and certain hydrophobic carriers may contain small amounts of water dissolved therein. Generally, compositions as disclosed herein that are “substantially free of water” contain, for example, less than about 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, 0.1%, 0.05% or 0.01% water on a weight/weight basis of the total weight of the carrier component of the composition. The compositions that still contain small quantities of water do not contain a sufficient amount of water such that an emulsion would be formed. As used herein, an “emulsion” refers to a mixture of two or more liquids that are normally immiscible wherein droplets of one liquid are dispersed in the other. As an example, a hydrophobic substance (e.g. oil) and an aqueous substance (e.g. water) are immiscible liquids that may form an emulsion when droplets of one are dispersed in the other. A dispersion of water droplets in oil is a water-in-oil (W/O) emulsion in which the water (aqueous phase) forms a discontinuous phase and the oil (hydrophobic phase) forms a continuous phase. “Water-in-oil emulsion” or “W/O”, as used herein, refers to an emulsion of a hydrophobic phase in an aqueous phase. A dispersion of oil droplets in water is an oil-in-water (O/W) emulsion in which the oil (hydrophobic phase) forms a discontinuous phase and the water (aqueous phase) forms a continuous phase. “Oil-in-water emulsion” or “O/W”, as used herein, refers to an emulsion of a hydrophobic phase in an aqueous phase. “Conventional emulsion”, as used herein, refers to emulsions are composed of numerous emulsifier-coated fluid droplets dispersed within another immiscible fluid medium, wherein the emulsifier forms an interfacial thin layer surrounding individual droplets that create interactions with hydrophilic and hydrophobic phases.
[0071] Lipids [0072] The pharmaceutical compositions and dried preparations of the present invention comprise at least lipid. In some embodiments, the lipid is an amphipathic lipid or a mixture of amphipathic lipids. Although any amphipathic lipid may be used, particularly suitable lipids may include those with at least one fatty acid chain containing at least 4 carbons, and typically about 4 to 28 carbons in length. The fatty acid chain may contain any number of saturated and/or unsaturated bonds. There are numerous amphipathic lipids which may be used in the pharmaceutical compositions of the present invention, and the pharmaceutical compositions of the present invention may contain a single type of amphipathic lipid or a mixture of different types of amphipathic lipids. The amphipathic lipid may be a natural lipid or a synthetic lipid. Non-limiting examples of amphipathic lipids for use in the present invention include phospholipids, sphingolipids, sphingomyelin, cerobrocides, gangliosides, ether lipids, sterols, cholesterol, glycerophospholipid, cardiolipin, cationic lipids, anionic lipids, and lipids modified with poly (ethylene glycol) and other polymers. Synthetic lipids may include, without limitation, the following fatty acid constituents: lauroyl, myristoyl, palmitoyl, stearoyl, arachidoyl, oleoyl, linoleoyl, erucoyl, or combinations of these fatty acids. An “amphipathic lipid” is a lipid having both hydrophilic and hydrophobic parts or characteristics. The term “amphipathic” may be used interchangeably with “amphiphile” or “amphiphilic”. The hydrophobic portion of an amphipathic lipid may be a large hydrocarbon moiety, either linear or cyclic, such as a long chain of the form CH3(CH2)n, with n > 4. The hydrophilic portion of an amphipathic lipid may be either a charged group or a polar uncharged group. Charged groups include anionic and cationic groups. Examples of anionic charged groups include the following (wherein the hydrophobic part of the molecule is represented by "R"): carboxylates: RCO2 ̄ ; sulfates: RSO4 ̄ ; sulfonates: RSO3 ̄ ; and phosphates (the charged functionality in phospholipids). Cationic charged groups include, for example, amines: RNH3+ ("R" again representing the hydrophobic part of the molecule). Uncharged polar groups include, for example, alcohols with large R groups, such as diacyl glycerol (DAG). Amphipathic lipids may have several hydrophobic parts, several hydrophilic parts, or several of both. Cholesterol is also an amphiphilic lipid. [0073] In some embodiments, the lipid is a phospholipid or a mixture of phospholipids. Broadly defined, a “phospholipid” is a member of a group of lipid compounds that yield on hydrolysis phosphoric acid, an alcohol, fatty acid, and nitrogenous base. Phospholipids that may be used include, for example and without limitation, those with at least one head group selected from the group consisting of phosphoglycerol, phosphoethanolamine, phosphoserine, phosphocholine (for example DOPC; 1,2-Dioleoyl-sn-glycero-3-phosphocholine) and
phosphoinositol. In some embodiments, the phospholipid may be phosphatidylcholine or a mixture of lipids comprising phosphatidylcholine. In some embodiments, the phospholipid may be DOPC or a mixture of phospholipids such as a lecithin (for example Lipoid S100 lecitihin). Lecithin is a mixture of phospholipids typically derived from biological sources such as eggs, soybean, and other vegetable sources. In some embodiments, the phospholipid is sphingomyelin. Sphingomyelin contains sphingosine, an amino alcohol with a long unsaturated hydrocarbon chain. A fatty acyl side chain is linked to the amino group of sphingosine by an amide bond, to form ceramide. The hydroxyl group of sphingosine is esterified to phosphocholine. [0074] In some embodiments, the at least one lipid is present in the pharmaceutical composition at a concentration of 60-160 mg/mL such as, for example 132 mg/mL. In some embodiments, the at least one lipid is present in the pharmaceutical composition at a concentration of at least 60 mg/mL, at least 70 mg/mL, at least 80 mg/mL, at least 90 mg/mL, at least 100 mg/mL, at least 110 mg/mL, at least 120 mg/mL, at least 132 mg/mL, at least 140 mg/mL, at least 150 mg/mL, or at least 160 mg/mL. [0075] In some embodiments, the at least one lipid is a mixture of a phospholipid and a sterol such as cholesterol. In some embodiments, the at least one lipid is a mixture of DOPC and cholesterol. In some embodiments, the at least one lipid is a mixture of lecithin and cholesterol. In some embodiments, the cholesterol or other sterol is used in an amount equivalent to about 2%, 5%, 10%, 12%, 15%, or 20% of the weight of phospholipid or mixture of phospholipids. In some embodiments, the amount of cholesterol or other sterol used is determined as the amount to sufficiently stabilize the formation of lipid-based structures formed by the phospholipid when suspended in the hydrophobic carrier. [0076] Lipid vesicle particles [0077] As used herein, the term “lipid vesicle particle” may be used interchangeably with “lipid vesicle”. A lipid vesicle particle refers to a complex or structure having an internal environment separated from the external environment by a continuous layer of enveloping lipids. In the context of the present disclosure, the expression “layer of enveloping lipids” can mean a single layer lipid membrane (e.g. as found on a micelle or reverse micelle), a bilayer lipid membrane (e.g. as found on a liposome) or any multilayer membrane formed from single and/or bilayer lipid membranes. The layer of enveloping lipids is typically a single layer, bilayer or multilayer throughout its circumference, but it is contemplated that other conformations may be possible such that the layer has different configurations over its circumference. The lipid vesicle particle may contain, within its internal environment, other vesicle structures (i.e. it may be
multivesicular). The term “lipid vesicle particle” encompasses many different types of structures, including without limitation micelles, reverse micelles, unilamellar liposomes, multilamellar liposomes and multivesicular liposomes. [0078] The lipid vesicle particles may take on various different shapes, and the shape may change at any given time (e.g. upon drying, sizing, or mixing with therapeutic agents). Typically, lipid vesicle particles are spherical or substantially spherical structures. By “substantially spherical” it is meant that the lipid vesicles are close to spherical, but may not be a perfect sphere. Other shapes of the lipid vesicle particles include, without limitation, oval, oblong, square, rectangular, triangular, cuboid, crescent, diamond, cylinder or hemisphere shapes. Any regular or irregular shape may be formed. Further, a single lipid vesicle particle may comprise different shapes if it is multivesicular. For example, the outer vesicle shape may be oblong or rectangular while an inner vesicle may be spherical. Exemplary embodiments of lipid vesicle particles include, without limitation, single layer vesicular structures (e.g. micelles or reverse micelles) and bilayer vesicular structures (e.g. unilamellar or multilamellar vesicles), or various combinations thereof. [0079] By “single layer” it is meant that the lipids do not form a bilayer, but rather remain in a layer with the hydrophobic part oriented on one side and the hydrophilic part oriented on the opposite side. By “bilayer” it is meant that the lipids form a two-layered sheet, such as with the hydrophobic part of each layer internally oriented toward the center of the bilayer with the hydrophilic part externally oriented. Alternatively, the opposite configuration is also possible, i.e. with the hydrophilic part of each layer internally oriented toward the center of the bilayer with the hydrophobic part externally oriented. The term “multilayer” is meant to encompass any combination of single and bilayer structures. The form adopted may depend upon the specific lipid that is used. [0080] The lipid vesicle particles may be formed from single layer lipid membranes, bilayer lipid membranes and/or multilayer lipid membranes. The lipid membranes are predominantly comprised of and formed by lipids, but may also comprise additional components. For example, and without limitation, the lipid membrane may include stabilizing molecules to aid in maintaining the integrity of the structure. Any available stabilizing molecule may be used such as, for example, a sterol such as cholesterol. [0081] The lipid vesicle particles may be formed by lipids, such as amphipathic lipids, as disclosed herein.
[0082] Lipid vesicle particles may be comprised of a single layer lipid assembly. There are various types of these lipid vesicle particles which may form, and the compositions disclosed herein may comprise a single type of lipid vesicle particle having a single layer lipid assembly or comprise a mixture of different such lipid vesicle particles. Lipid vesicle particles having a single layer lipid assembly may comprise aggregates of lipids with the hydrophobic part of the lipids oriented outwards toward the hydrophobic carrier and the hydrophilic part of the lipids aggregating as a core. These structures do not necessarily form a continuous lipid layer membrane. Lipid vesicle particles having a single layer lipid assembly comprise reverse micelles. A typical micelle in aqueous solution forms an aggregate with the hydrophilic parts in contact with the surrounding aqueous solution, sequestering the hydrophobic parts in the micelle center. In contrast, in a hydrophobic carrier, an inverse/reverse micelle forms with the hydrophobic parts in contact with the surrounding hydrophobic solution, sequestering the hydrophilic parts in the micelle center. The size of the lipid-based structures having a single layer lipid assembly is in the range of from 2 nm (20 A) to 20 nm (200 A) in diameter. [0083] Lipid vesicle particles may be bilayer vesicular structures, such as for example, a liposome. Liposomes are completely closed lipid bilayer membranes. Liposomes may be unilamellar vesicles (possessing a single bilayer membrane), multilamellar vesicles (characterized by multimembrane bilayers whereby each bilayer may or may not be separated from the next by an aqueous layer) or multivesicular vesicles (possessing one or more vesicles within a vesicle). Liposomes form in aqueous or partially aqueous environments, and may form when the compositions herein are not water-free or may form in the aqueous solvents and solutions used in the preparation of pharmaceutical compositions. [0084] In some embodiments, the lipid vesicle particles are liposomes. In an embodiment, the liposomes are unilamellar, multilamellar, multivesicular or a mixture thereof. In an embodiment, the mean particle size of the liposomes is ≥80 nm. Thus, in an embodiment, the mean particle size of the liposomes used in the methods disclosed herein is in the range of 80 nm to 120 nm, with a PDI of ≤0.1. As used herein, “mean” refers to the arithmetic mean of the particle size of the lipid vesicle particles in a given population. It is a synonym for average. As such, “mean particle size” is intended to refer to the sum of the diameters of each lipid vesicle particle of a population, divided by the total number of lipid vesicle particles in the population (e.g. in a population with 4 lipid vesicle particles with particle sizes of 95 nm, 98 nm, 102 nm and 99 nm, the mean particle size is (95+98+102+99)/4 = 98.5 nm). However, as the skilled person will appreciate, lipid vesicle particles may not be perfectly spherical, and therefore the “particle size” of a given vesicle particle may not be an exact measure of its diameter. Rather,
the particle size may be defined by other means known in the art, including for example: the diameter of the sphere of equal area or the largest perpendicular distance between parallel tangents touching opposite sides of the particle (Feret’s statistical diameter). [0085] In some embodiments, the sized lipid vesicle particles have a mean particle size of less than or equal to 120 nanometers (i.e. ≤120 nm) and a polydispersity index (PDI) of ≤0.1. In an embodiment, the sized lipid vesicle particles have a mean particle size of ≤115 nm, more particularly still ≤110 nm and more particularly still ≤100 nm. In an embodiment, the mean particle size of the sized lipid vesicle particles is between 50 nm and 120 nm. In an embodiment, the mean particle size of the sized lipid vesicle particles is between 80 nm and 120 nm. In an embodiment, the mean particle size of the sized lipid vesicle particles is between about 80 nm and about 115 nm, about 85 nm and about 115 nm, about 90 nm and about 115 nm, about 95 nm and about 115 nm, about 100 nm and about 115 nm or about 105 nm and about 115 nm. In an embodiment, the mean particle size of the sized lipid vesicle particles is about 80 nm, about 81 nm, about 82 nm, about 83 nm, about 84 nm, about 85 nm, about 86 nm, about 87 nm, about 88 nm, about 89 nm, about 90 nm, about 91 nm, about 92 nm, about 93 nm, about 94 nm, about 95 nm, about 96 nm, about 97 nm, about 98 nm, about 99 nm, about 100 nm, about 101 nm, about 102 nm, about 103 nm, about 104 nm, about 105 nm, about 106 nm, about 107 nm, about 108 nm, about 109 nm, about 110 nm, about 111 nm, about 112 nm, about 113 nm, about 114 nm, about 115 nm, about 116 nm, about 117 nm, about 118 nm or about 119 nm. In an embodiment, the mean particle size is 120 nm. [0086] As used herein, polydispersity index (PDI) is a measure of the size distribution of the lipid vesicle particles. It is known in the art that the term “polydispersity” may be used interchangeably with “dispersity”. The PDI can be calculated by determining the mean particle size of the lipid vesicle particles and the standard deviation from that size. There are techniques and instruments available for measuring the PDI of lipid vesicle particles. For example, DLS is a well-established technique for measuring the particle size and size distribution of particles in the submicron size range, with available technology to measure particle sizes of less than 1 nm (LS Instruments, CH; Malvern Instruments, UK). [0087] Lipid vesicle particles having a mean particle size of ≤120 nm and a PDI of ≤0.1 may be prepared and provided by any suitable means. In an embodiment, the lipid vesicle particles are prepared in a manner in which their size is controlled in order to achieve the mean particle size of ≤120 nm and a PDI of ≤0.1. In an embodiment, lipid vesicle particles are subjected to one or more sizing steps or protocols to achieve the mean particle size of ≤120 nm
and a PDI of ≤0.1. In an embodiment, the lipid vesicle particles may be prepared and provided by any combination of controlling their size during manufacture, performing sizing steps and/or any other means available in the art. In an embodiment, the lipid vesicle particles must be subjected to one or more active steps of sizing in order to achieve the mean particle size of ≤120 nm and a PDI of ≤0.1. In an embodiment, the sizing is performed by filter-extrusion whereby lipid vesicle particles are passed through a filter membrane or a series of filter membranes (e.g. polycarbonate membranes) of appropriate pore size. As such, as used herein, “sized lipid vesicle particles” refers to lipid vesicle particles that have been prepared by a means in which their size is controlled to attain a mean particle size of ≤120 nm and a PDI of ≤0.1 and/or they are sized to meet the criteria of having a mean particle size of ≤120 nm and a PDI of ≤0.1. The skilled person will be well aware of techniques available for providing lipid vesicle particles having a mean particle size of ≤120 nm and a PDI of ≤0.1. Reference herein to “non- sized lipid vesicle particles” or a “non-sized lipid vesicle particle preparation” means that the lipid vesicle particles have not be subject to procedures that limit their size to meet the defined size criteria, and/or they do not have a mean particle size of ≤120 nm and a PDI of ≤0.1. [0088] In an embodiment, the sized lipid vesicle particles may be prepared from a lipid precursor that naturally forms lipid vesicle particles of the required size. For example, and without limitation, the sized lipid vesicle particles may be prepared using Presome® (Nippon Fine Chemical, Japan). Presome® is a dry powder precursor made up of different lipid combinations. Presome® is supplied ready to be wetted in a suitable buffer to prepare liposomes. The liposomes formed from Presome® have an average particle size of about 93 nm, and sizing procedures (e.g. membrane extrusion, high pressure homogenization, etc.) can be used to achieve the required mean particle size of ≤120 nm and PDI of ≤0.1. In an embodiment, Presome® may for example be wetted in sodium acetate, pH 9.0 ± 0.5 to form liposomes. In an embodiment, the Presome® bulk dry powder may be made from DOPC/cholesterol (10:1 (w/w)) or DOPC alone. [0089] In another embodiment, standard procedures for preparing lipid vesicle particles of any size may be employed. For example, conventional liposome forming processes may be used, such as the hydration of solvent-solubilized lipids. Exemplary methods of preparing liposomes are discussed, for example, in Gregoriadis 1990; and Frezard 1999. After the lipid vesicle particles are prepared, the non-sized lipid vesicle particle preparation is subjected to a sizing procedure to obtain lipid vesicle particles having a mean particle size of ≤120 nm and a PDI of ≤0.1. There are various techniques available for sizing lipid vesicle particles (see e.g.
Akbarzadeh 2013). For example, in an embodiment, the non-sized lipid vesicle particle preparation may be sized by high pressure homogenization (microfluidizers), sonication or membrane based extrusion. [0090] In an embodiment, the sizing of the non-sized lipid vesicle particle preparation is performed using high pressure homogenization to obtain sized lipid vesicle particles having a mean particle size of >120 nm and a PDI of >0.1, and the lipid vesicle particles obtained via high pressure homogenization can then be further sized down using membrane based extrusion. In an embodiment, membrane based extrusion is performed by passing the sized lipid vesicle particle preparation 5-20 times through a 0.1 um polycarbonate membrane or, alternatively, 5-20 times through a 0.08 um polycarbonate membrane, thereby attaining a mean particle size of ≤120 nm and PDI of ≤0.1. [0091] In an embodiment, the sized lipid vesicle particles may be prepared by adding the lipids to a suitable solvent (e.g. sodium phosphate, 50 mM, pH 7.0), shaking and/or stirring the lipid mixture (e.g. at 300 RPM for about 1 hour) and using membrane based extrusion to obtain the sized lipid vesicle particles. Exemplary, non-limiting embodiments of membrane based extrusion include: (i) passing a non-sized lipid vesicle particle preparation 20-40 times through a 0.2 μm polycarbonate membrane, and then 10-20 times through a 0.1 μm polycarbonate membrane; or (ii) passing a non-sized lipid vesicle particle preparation 20-40 times through a 0.2 μm polycarbonate membrane, then 10-20 times through a 0.1 μm polycarbonate membrane, and then 10-20 times through a 0.08 μm polycarbonate membrane. [0092] In a particular embodiment, the sizing may be performed by passing a non-sized lipid vesicle particle preparation 25 times through a 0.2 μm polycarbonate membrane, and then 10 times through a 0.1 μm polycarbonate membrane. In another particular embodiment, the sizing may be performed by passing a non-sized lipid vesicle particle preparation 25 times through a 0.2 μm polycarbonate membrane, then 10 times through a 0.1 μm polycarbonate membrane, and then 15 times through a 0.08 μm polycarbonate membrane. [0093] Dried preparation [0094] As used herein, the term “dried preparation” refers to a mixture of components as described herein that is dried by a technique known in the art in order to remove all or substantially all of a solvent. The term “dried preparation” does not necessarily mean that the preparation is completely dry. For example, depending on the solvent or solvents used in the methods disclosed herein, it is possible that a small component of volatile and/or non-volatile
material will remain in the dried preparation. In an embodiment, the non-volatile material will remain. By “dried preparation”, it is meant that the preparation no longer contains substantial quantities of water and/or organic solvent. The process used to dry the preparation should be capable of removing substantially all water and/or organic solvent. Thus, in an embodiment, the dried preparation is completely free of water and/or organic solvent. In another embodiment, the dried preparation may contain a residual moisture content based on the limitations of the drying process (e.g. lyophilization). This residual moisture content will typically be less than 2%, less than 1%, less than 0.5%, less than 0.25%, less than 0.1%, less than 0.05% or less by weight of the dried preparation. This residual moisture content will not be more than 5% by weight of the dried preparation as this would result in a product that is not clear. [0095] Various methods may be used to dry compositions are known in the art. In an embodiment, the drying is performed by lyophilization, spray freeze-drying, or spray drying. The skilled person is well-aware of these drying techniques and how they may be performed. In an embodiment, the drying is performed by lyophilization. As used herein, “lyophilization”, “lyophilized” and “freeze-drying” are used interchangeably. As is well known in the art, lyophilization works by freezing the material and then reducing the surrounding pressure to allow the volatile solvent (e.g. water) in the material to sublime directly from the solid phase to the gas phase. Any conventional freeze-drying procedure may be used to carry out the drying step of the methods disclosed herein. In an embodiment, the lyophilization is performed by sequential steps of loading, freezing, evacuation and drying (e.g. primary drying and secondary drying). In an embodiment, lyophilization of the sized lipid vesicle particle/therapeutic agent mixture can be performed within a sealed bag in a benchtop freeze dryer. This may be particularly advantageous because it reduces the number of steps that must be performed in a sterile laboratory environment and allows for the rapid manufacture of smaller batch sizes. For example, after sterile filtration of the sized lipid vesicle particle/therapeutic agent mixture, aseptically filled vials containing the mixture can be loaded and sealed within a sterile bag under sterile conditions. These sterile, sealed units can then undergo lyophilization in an open laboratory (i.e. non-sterile environment) using a benchtop freeze dryer. By this method, it is also possible to perform the freeze-drying with multiple different sealed units in a single freeze dryer. This may reduce the cost and time of manufacture by avoiding expensive freeze-drying steps in sterile laboratory environments using large-scale freeze dryers. Also, multiple different small- scale batches of dried lipid/therapeutic agent preparation may be prepared simultaneously in separate sealed sterile bags. The lyophilization may be performed in any suitable freeze dryer. In an embodiment, the freeze dryer is a benchtop freeze dryer. In an embodiment, the freeze dryer
is a Virtis benchtop lyophilizer. In an embodiment, the freeze dryer is in an open laboratory (i.e. non-sterile environment). [0096] The methods disclosed herein may further comprise a step of sterilization. Sterilization may be performed by any method known in the art. In an embodiment, the sterilization is performed by sterile filtration, steam heat sterilization, irradiation (e.g. gamma irradiation) or chemical sterilization. In a particular embodiment, the sterilization is performed by sterile filtration. In an embodiment, the sterile filtration is be performed before drying. Any conventional procedure for sterile filtration may be employed so long as it does not affect the solubility and stability of the therapeutic agents, lipid-based adjuvant, and/or polyI:C polynucleotide in the sized lipid vesicle particle/therapeutic agent mixture. In this regard, it may be desirable to perform the sterile filtration under low pressure conditions (e.g. between 30-50 psi). The serial filtration may be performed using commercially available sterile filtration membranes (e.g. MilliporeSigma). In an embodiment, the sterile filtration is performed using a 0.22 μm-rated membrane, a 0.2 μm-rated membrane and/or a 0.1 μm-rated membrane. In an embodiment, the sterile filtration is performed by a single passage of the mixture through a single filtration membrane. In another embodiment, the sterile filtration is performed by serially passing the mixture sequentially through a combination of the same or different sized filtration membranes. [0097] The methods disclosed herein may further comprise a step of confirming that the sized lipid vesicle particles have retained a mean particle size of ≤120 nm and PDI of ≤0.1. As described elsewhere herein, there are several techniques, instruments and services that are available to measure the mean particle size and PDI of lipid vesicle particles, such as for example and without limitation TEM, SEM, AFM, FTIR, XPS, XRD, MALDI-TOF-MS, NMR and DLS. In an embodiment, the step of confirming the size and PDI of the lipid vesicle particles is performed using a DLS ZETASIZER NANO-S particle size analyzer. [0098] The methods disclosed herein may further comprise a step of evaluating the stability of the lipids, therapeutic agent(s), lipid-based adjuvant, and/or polyI:C polynucleotide before and/or after the drying step. The stability of the components may be measured by any known means or method. For example and without limitation, stability of the dried preparation may be determined by the appearance of the dried preparation (lyophilisate) or measurement of the content of the components over time (e.g. by HPLC, RP-HPLC, IEX-HPLC, etc.). HPLC is a technique which can be used to separate, identify and quantify each component in a mixture. Thus, by using HPLC, RP-HPLC or IEX-HPLC it is possible to determine the approximate
quantity of the lipids, therapeutic agents and other components, as well as characterize the components qualitatively (e.g. observe impurities, degradation products, etc.). In other embodiments, stability may evaluated upon solubilization in a hydrophobic carrier by various methods, such as for example: appearance of the solubilized product; identification and quantification of lipids, therapeutic agent(s), lipid-based adjuvant, and/or polyI:C polynucleotide, impurities or degradants (e.g. by RP-HPLC, IEX-HPLC, etc.); particle size of the lipid vesicle particles (e.g. by SAXS); optical density; viscosity (e.g. as per Ph.Eur.2.2.9); pH; extractable volume, such as from a syringe (e.g. as per Ph.Eur.2.9.17), and immunogenicity assays (e.g. ELISpot). [0099] In embodiments of the methods disclosed herein, lipids may first be dissolved and mixed in an organic solvent. In embodiments where different types of lipid are used, this step will allow a homogenous mixture of the lipids to be formed. In an embodiment, these steps may be carried out in chloroform, chloroform:methanol mixtures, tertiary butanol or cyclohexane. In an embodiment, the lipids are prepared at 10-20mg lipid/mL organic solvent; however, higher or lower concentrations may also be used. In some embodiments, after mixing, the organic solvent is removed (e.g. by evaporation) to yield a lipid film. The lipid film may then be frozen and lyophilized to yield a dry lipid film. The dry lipid film may then be hydrated with an aqueous solution containing therapeutic agents/polyI:C polynucleotide/lipid-based adjuvant to provide a non-sized lipid vesicle particle preparation. The step of hydration may be performed with shaking and/or mixing (e.g. at 300 RPM for about 1 hour). In yet another embodiment of the methods disclosed herein, an aqueous solution of lipids may be combined with at least one solution containing therapeutic agents/polyI:C polynucleotide/lipid-based adjuvant. In another embodiment, one or more dry therapeutic agents/polyI:C polynucleotide/lipid-based adjuvant may be added to, and solubilized in, the aqueous solution of lipids or sized lipid vesicle preparation. These embodiments may be performed with shaking and/or mixing (e.g. at 300 RPM for about 1 hour). The above procedures are exemplary methods for providing a non-sized lipid vesicle particle preparation comprising non-sized lipid vesicles and one or more first therapeutic agents and/or polyI:C polylnucleotide. [0100] In some embodiments, therapeutic agents/polyI:C polynucleotide/lipid-based adjuvant are either solubilized in a solvent (e.g. aqueous or organic) prior to mixing with lipid vesicle particles or therapeutic agents/polyI:C polynucleotide/lipid-based adjuvant are solubilized upon being mixed with the lipid vesicle particles. In this latter embodiment, therapeutic agents/polyI:C polynucleotide/lipid-based adjuvant may be added as a dry powder to
a solution containing lipid vesicle particles or both the lipid vesicle particles and therapeutic agents/polyI:C polynucleotide/lipid-based adjuvant may be mixed together in a fresh solvent. [0101] Methods and uses [0102] The pharmaceutical compositions disclosed herein may find application in any instance in which it is desired to administer therapeutic agents to a subject. The subject may be a vertebrate, such as a fish, bird or mammal. In an embodiment, the subject is a mammal. In an embodiment, the subject is a human. In an embodiment, the pharmaceutical compositions may be used in methods for treating, preventing or diagnosing a disease, disorder or condition to which the therapeutic agent is targeted. In an embodiment, the pharmaceutical compositions may be used in methods for delivering a therapeutic agent to a subject. In an embodiment, the methods comprise administering to a subject the pharmaceutical composition as described herein. In an embodiment, the pharmaceutical compositions may be used in methods for modulating an immune response in a subject. As used herein, the term “modulating” is intended to refer to both immunostimulation (e.g. inducing or enhancing an immune response) and immunosuppression (e.g. preventing or decreasing an immune response). Typically, the method would involve one or the other of immunostimulation or immunosuppression, but it is possible that the method could be directed to both. As referred to herein, the “immune response” may either be a cell-mediated (CTL) immune response or an antibody (humoral) immune response. [0103] In some embodiments, the pharmaceutical compositions disclosed herein may be used for inducing a cell-mediated immune response to the therapeutic agents (e.g. antigens). As used herein, to “induce” an immune response is to elicit and/or potentiate an immune response. Inducing an immune response encompasses instances where the immune response is initiated, enhanced, elevated, improved or strengthened to the benefit of the host relative to the prior immune response status, for example, before the administration of a composition disclosed herein. [0104] In an embodiment, the pharmaceutical compositions disclosed herein may be used for inducing an antibody immune response to the therapeutic agents (e.g. antigens). An “antibody immune response” or “humoral immune response” (used interchangeably herein), as opposed to cell-mediated immunity, is mediated by secreted antibodies which are produced in the cells of the B lymphocyte lineage (B cells). Such secreted antibodies bind to antigens, such as for example those on the surfaces of foreign substances, pathogens (e.g. viruses, bacteria, etc.) and/or cancer cells, and flag them for destruction.
[0105] The pharmaceutical compositions disclosed herein may be useful for treating or preventing diseases and/or disorders ameliorated by a cell-mediated immune response or a humoral immune response. The pharmaceutical compositions disclosed herein may find application in any instance in which it is desired to administer therapeutic agents (e.g. antigens) to a subject to induce a cell-mediated immune response or a humoral immune response. In an embodiment, the pharmaceutical compositions is for inducing an antibody immune response and/or cell-mediated immune response to the therapeutic agents (e.g. antigens) in said subject. In an embodiment, pharmaceutical compositions is for the treatment and/or prevention of an infectious disease or cancer. [0106] “Treating” or “treatment of”, or “preventing” or “prevention of”, as used herein, refers to an approach for obtaining beneficial or desired results. Beneficial or desired results can include, but are not limited to, alleviation or amelioration of one or more symptoms or conditions, diminishment of extent of disease, stabilisation of the state of disease, prevention of development of disease, prevention of spread of disease, delay or slowing of disease progression (e.g. suppression), delay or slowing of disease onset, conferring protective immunity against a disease-causing agent and amelioration or palliation of the disease state. “Treating” or “preventing” can also mean prolonging survival of a patient beyond that expected in the absence of treatment and can also mean inhibiting the progression of disease temporarily or preventing the occurrence of disease, such as by preventing infection in a subject. “Treating” or “preventing” may also refer to a reduction in the size of a tumor mass, reduction in tumor aggressiveness, etc. Treating” may be distinguished from “preventing” in that “treating” typically occurs in a subject who already has a disease or disorder, or is known to have already been exposed to an infectious agent, whereas “preventing” typically occurs in a subject who does not have a disease or disorder, or is not known to have been exposed to an infectious agent. As will be appreciated, there may be overlap in treatment and prevention. For example, it is possible to be “treating” a disease in a subject, while at same time “preventing” symptoms or progression of the disease. Moreover, at least in the context of vaccination, “treating” and “preventing” may overlap in that the treatment of a subject is to induce an immune response that may have the subsequent effect of preventing infection by a pathogen or preventing the underlying disease or symptoms caused by infection with the pathogen. These preventive aspects are encompassed herein by expressions such as “treatment of an infectious disease” or “treatment of cancer”. [0107] In an embodiment, the pharmaceutical compositions disclosed herein may be used for treating and/or preventing an infectious disease, such as caused by a viral infection, in a
subject in need thereof. The subject may be infected with a virus or may be at risk of developing a viral infection. Viral infections that may be treated and/or prevented by the use or administration of a pharmaceutical composition as disclosed herein, without limitation, Cowpoxvirus, Vaccinia virus, Pseudocowpox virus, Human herpesvirus 1 , Human herpesvirus 2, Cytomegalovirus, Human adenovirus A-F, Polyomavirus, Human papillomavirus (HPV), Parvovirus, Hepatitis A virus, Hepatitis B virus, Hepatitis C virus, Human immunodeficiency virus, Orthoreovirus, Rotavirus, Ebola virus, parainfluenza virus, influenza A virus, influenza B virus, influenza C virus, Measles virus, Mumps virus, Rubella virus, Pneumovirus, respiratory syncytial virus (RSV), Rabies virus, California encephalitis virus, Japanese encephalitis virus, Hantaan virus, Lymphocytic choriomeningitis virus, Coronavirus (e.g. Sars-Cov-2), Enterovirus, Rhinovirus, Poliovirus, Norovirus, Flavivirus, Dengue virus, West Nile virus, Yellow fever virus and varicella. [0108] In an embodiment, the compositions disclosed herein may be used for treating and/or preventing an infectious disease, such as caused by a non-viral pathogen (such as a bacterium or protozoan) in a subject in need thereof. The subject may be infected with the pathogen or may be at risk of developing an infection by the pathogen. Without limitation, exemplary bacterial pathogens may include Anthrax (Bacillus anthracis), Brucella, Bordetella pertussis, Candida, Chlamydia pneumoniae, Chlamydia psittaci, Cholera, Clostridium botulinum, Coccidioides immitis, Cryptococcus, Diphtheria, Escherichia coli O157: H7, Enterohemorrhagic Escherichia coli, Enterotoxigenic Escherichia coli, Haemophilus influenzae, Helicobacter pylori, Legionella, Leptospira, Listeria, Meningococcus, Mycoplasma pneumoniae, Mycobacterium, Pertussis, Pneumonia, Salmonella, Shigella, Staphylococcus, Streptococcus pneumoniae and Yersinia enterocolitica. In a particular embodiment, the bacterial infection is Anthrax. Without limitation, exemplary protozoan pathogens may include those of the genus Plasmodium (Plasmodium falciparum, Plasmodium malariae, Plasmodium vivax, Plasmodium ovale or Plasmodium knowlesi), which cause malaria. [0109] In an embodiment, the pharmaceutical compositions disclosed herein may be for use in treating and/or preventing cancer in a subject in need thereof. The subject may have cancer or may be at risk of developing cancer. As used herein, the terms “cancer”, “cancer cells”, “tumor” and “tumor cells”, (used interchangeably) refer to cells that exhibit abnormal growth, characterized by a significant loss of control of cell proliferation or cells that have been immortalized. The term “cancer” or “tumor” includes metastatic as well as non-metastatic cancer or tumors. A cancer may be diagnosed using criteria generally accepted in the art, including the presence of a malignant tumor.
[0110] Without limitation, cancers that may be capable of being treated and/or prevented by the use or administration of a pharmaceutical composition as disclosed herein include carcinoma, adenocarcinoma, lymphoma, leukemia, sarcoma, blastoma, myeloma, and germ cell tumors. Without limitation, particularly suitable embodiments may include glioblastoma, multiple myeloma, ovarian cancer, breast cancer, fallopian tube cancer, prostate cancer or peritoneal cancer. In one embodiment, the cancer may be caused by a pathogen, such as a virus. Viruses linked to the development of cancer are known to the skilled person and include, but are not limited to, human papillomaviruses (HPV), John Cunningham virus (JCV), Human herpes virus 8, Epstein Barr Virus (EBV), Merkel cell polyomavirus, Hepatitis C Virus and Human T cell leukaemia virus-1. In an embodiment, the cancer is one that expresses one or more tumor- specific neoantigens. In a particular embodiment, the cancer is breast cancer, ovarian cancer, prostate cancer, fallopian tube cancer, peritoneal cancer, glioblastoma or diffuse large B cell lymphoma. [0111] The pharmaceutical compositions disclosed herein may be useful for either the treatment or prophylaxis of cancer; for example, a reduction of the severity of cancer (e.g. size of the tumor, aggressiveness and/or invasiveness, malignancy, etc.) or the prevention of cancer recurrences. [0112] The pharmaceutical composition as disclosed herein may be administered by any suitable route. In an embodiment, the route of administration is injection, such as subcutaneous injection. [0113] Kits [0114] The pharmaceutical compositions disclosed herein are optionally provided to a user as a kit comprising the individual components which may be assembled to produce the pharmaceutical composition. In an embodiment, the kit is for preparing a composition for the treatment, prevention and/or diagnosis of a disease, disorder or condition or for the delivery of a therapeutic agent. [0115] The kits can further comprise one or more additional reagents, packaging materials, and an instruction set or user manual detailing preferred methods of using the kit components. In an embodiment, the containers are vials. [0116] The invention is further illustrated by the following non-limiting examples. EXAMPLES
[0117] Example 1 [0118] First, a vial containing DPX-FP (IMV-Inc, Dartmouth, Canada) was reconstituted in 0.45 mL of Montanide ISA 51 oil diluent (SEPPIC, France) by soaking for 5 minutes and shaking for 2 minuntes, then vortexing for one minute. Next, 0.45 mL of this oil reconstituted DPX-FP product was then added to a vial containing DPX-PAM3CSK4 (IMV-Inc, Dartmouth, Canada) and reconstitution was completed by soaking for 5 minutes, shaking for 2 minutes, and vortexing for one minute to obtain the final concentrations of: FP peptide 9 ug/50 uL dose; DNA based polyI:C polynucleotide (dIdC) 18 ug/50 uL dose; PAM3CSK42 ug/50 uL dose; and Lipid 12.5 mg/50 uL dose. [0119] The DPX-FP product vial was previously prepared by adding FP (NeoMPS) and DNA based polyI:C polynucleotide (dIdC) stock (Biospring) to a lipid-mixture solution, mixing well and freeze-drying. A lipid-mixture (132 mg/mL) containing DOPC and cholesterol in a 10:1 ratio (w:w) (Lipoid GmBH, Germany) was dissolved in 40% tertiary-butanol by shaking well at 300 RPM at room temperature for 1 hour or until dissolved. Next, FP stock (10 mg/mL) was prepared in DMSO and DNA based polyI:C polynucleotide adjuvant stock (10 mg/mL) was prepared in sterile water. To a 0.5 mL aliquot of lipid-mixture solution, 10 µL of FP stock was added to obtain 0.1 mg/mL final fill concentration, shaken well at 300 RPM for 5 minutes. To the formed FP-lipid-mixture solution, 20 µL of DNA based polyI:C polynucleotide (dIdC) stock was added to obtain 0.2 mg/mL final fill concentration, shaken well at 300 RPM for 5 minutes. The volume was adjusted to 1.0 mL with 40% tertiary-butanol, freeze-dried, and stored at -20 C. The dried preparation is reconstituted in 0.45 mL oil diluent when needed. [0120] The DPX-PAM3CSK4 vial was prepared as follows: Briefly, a homogenous lipid- mixture of DOPC and cholesterol (Lipoid GmbH, Germany) was weighed to obtain 132 mg/mL of the lipid-mixture and was added to sodium phosphate, 100 mM, pH 6.0 with shaking at 300 RPM for about 1 hour. The mixture was then sized by passing the material 25 times through a 0.2 µm polycarbonate membrane, then 10 times through a 0.1 µm polycarbonate membrane, and then 15 times through a 0.08 µm polycarbonate membrane to attain a particle size of ≤100 nm with a polydispersity index (PDI) of ≤0.1. The lipopeptide adjuvant PAM3CSK4 (Polypeptide group, San Diego, USA) was then added at 0.02 mg/mL to the sized lipid vesicles with shaking at 300 RPM for about 15 minutes. The mixture was then sterile filtered, aseptically filled in 1.0 mL aliquots into sterile glass vials, freeze-dried, and stored at -20°C. The dried preparation is reconstituted in 0.45 mL oil diluent when needed. [0121] Example 2
[0122] First, a vial containing DPX-MVP-S (IMV-Inc, Dartmouth, Canada) was reconstituted in 0.7 mL of Montanide ISA 51 oil (SEPPIC, France) by soaking for 5 minutes, shaking for 2 mins, and vortexing for one minute. Next, 0.45 mL of this oil reconstituted DPX- MVP-S product was then added to a vial containing DPX-PAM3CSK4 (IMV-Inc, Dartmouth, Canada), and reconstitution was completed by soaking for 5 minutes, shaking for 2 minutes, and vortexing for one minute to obtain the final concentrations of: MVP-S peptide each at 45 ug/50 uL dose; T-helper peptide A16L at 22.5 ug/50 uL dose; DNA based polyI:C polynucleotide (dIdC) 18 ug/50 uL dose; PAM3CSK42 ug/50 uL dose; and Lipid 12.5 mg/50 uL dose. [0123] The DPX-MVP-S vial was prepared as follows: Briefly, DNA based polyI:C polynucleotide (dIdC) (BioSpring GmbH (Frankfurt, Germany) and peptide antigens (PolyPeptide Laboratories (San Diego, CA, USA) stock solutions were prepared as listed below: Stock# Component Solvent 5)
in the following order: (4), (2), (3), (5) and then (1). The pH was adjusted to 10.0 ± 0.5. A 10:1 (w:w), homogenous lipid-mixture of DOPC and cholesterol (Lipoid GmbH, Germany) was weighed to obtain 132 mg/mL of the lipid-mixture and added to the peptide/polynucleotide
solution to form an intermediate bulk (non-sized) and mixed using a Silverson high speed mixer. The pH was adjusted to 10.0 ± 0.5, if required. The intermediate bulk was then sized using an Emulsiflex C5 or C55 extruder by passing the material 35 times through a 0.2 µm polycarbonate membrane and then 10 times through a 0.1 µm polycarbonate membrane to attain a particle size of ≤120 nm with a PDI of ≤0.1. The peptide stock solutions (6) and (7) were then added to the sized lipid vesicle particle bulk immediately after preparation. The final pH of the solution was adjusted to 7.0 ± 0.5. The final preparation was then sterile filtered, aseptically filled (1.6 mL aliquots) into sterile glass vials, freeze-dried, and stored at -20°C. The dried preparation is reconstituted in 0.7 mL oil diluent when needed. [0125] The DPX-PAM3CSK4 vial was prepared as follows: Briefly, homogenous lipid- mixture of DOPC and cholesterol (Lipoid GmbH, Germany) was weighed to obtain 132 mg/mL of the lipid-mixture was added to sodium phosphate, 100 mM, pH 6.0 with shaking at 300 RPM for about 1 hour. The mixture was then sized by passing the material 25 times through a 0.2 µm polycarbonate membrane, then 10 times through a 0.1 µm polycarbonate membrane, and then 15 times through a 0.08 µm polycarbonate membrane to attain a particle size of ≤100 nm with a PDI of ≤0.1. The lipopeptide adjuvant PAM3CSK4 (Polypeptide group, San Diego, USA) was then added at 0.02 mg/mL to the sized lipid vesicles with shaking at 300 RPM for about 15 minutes. The mixture was then sterile filtered, aseptically filled to 1.0 mL aliquots into sterile glass vials, freeze-dried, and stored at -20°C. The dried preparation is reconstituted in 0.45 mL oil diluent when needed.
Claims
CLAIMS: 1. A method for preparing a pharmaceutical composition, comprising: (a) providing: (i) a first dried preparation comprising at least one therapeutic agent, a lipid, and a polyI:C polynucleotide, and (ii) a second dried preparation comprising a lipid-based adjuvant and a lipid; (b) combining the first dried preparation with a hydrophobic carrier to produce a reconstituted preparation; and (c) combining the reconstituted preparation with the second dried preparation to produce a pharmaceutical composition.
2. A method for preparing a pharmaceutical composition, comprising: (a) providing: (i) a first dried preparation comprising at least one therapeutic agent, a lipid, and a polyI:C polynucleotide, and (ii) a second dried preparation comprising a lipid-based adjuvant and a lipid; (b) combining the second dried preparation with a hydrophobic carrier to produce a reconstituted preparation; and (c) combining the reconstituted preparation with the first dried preparation to produce a pharmaceutical composition.
3. The method of claim 1 or 2, wherein providing the first dried preparation comprises: (i) providing lipid vesicle particles having a mean particle size of ≤120 nm and a polydispersity index (PDI) of ≤0.1; (ii) mixing the lipid vesicle particles with at least one solubilized therapeutic agent and a polyI:C polynucleotide to form a mixture; and (iii) drying the mixture formed in step (ii) to form a first dried preparation comprising a therapeutic agent, a lipid, and a polyI:C polynucleotide.
4. The method of claim 3, wherein step (i) comprises sizing lipid vesicle particles to provide the lipid vesicle particles having a mean particle size of ≤120 nm and a PDI of ≤0.1.
5. The method of claim 4, wherein the sizing is by filter-extrusion.
6. The method of claim 4 or 5, wherein the sizing is by extrusion through one or more polycarbonate membranes, such as a 0.2 µm polycarbonate membrane, a 0.1 µm polycarbonate membrane and/or a 0.08 µm polycarbonate membrane.
7. The method of any one of claims 4 to 6, wherein the sizing is by extrusion (i) 20-40 times through a 0.2 μm polycarbonate membrane, and then 10-20 times through a 0.1 μm polycarbonate membrane; or (ii) 20-40 times through a 0.2 μm polycarbonate membrane, then 10-20 times through a 0.1 μm polycarbonate membrane, and then 10-20 times through a 0.08 μm polycarbonate membrane.
8. The method of any one of claims 4 to 7, wherein the sizing is by extrusion (i) 25 times through a 0.2 μm polycarbonate membrane, and then 10 times through a 0.1 μm polycarbonate membrane or (ii) 25 times through a 0.2 μm polycarbonate membrane, then 10 times through a 0.1 μm polycarbonate membrane, and then 15 times through a 0.08 μm polycarbonate membrane.
9. The method of claim 4, wherein the sizing is performed by first using high pressure homogenization to obtain sized lipid vesicle particles having a mean particle size of >120 nm and a PDI of >0.1 and then sizing by extrusion (i) 5-20 times through a 0.1 μm polycarbonate membrane or (ii) 5-20 times through a 0.08 μm polycarbonate membrane.
10. The method of any one of claims 3 to 9, wherein the mean particle size of the lipid vesicle particles is between about 80 nm and about 120 nm.
11. The method of any one of claims 3 to 10, wherein the mean particle size of the lipid vesicle particles is about 80 nm, about 81 nm, about 82 nm, about 83 nm, about 84 nm, about 85 nm, about 86 nm, about 87 nm, about 88 nm, about 89 nm, about 90 nm, about 91 nm, about 92 nm, about 93 nm, about 94 nm, about 95 nm, about 96 nm, about 97 nm, about 98 nm, about 99 nm, about 100 nm, about 101 nm, about 102 nm, about 103 nm, about 104 nm, about 105 nm, about 106 nm, about 107 nm, about 108 nm, about 109 nm, about 110 nm, about 111 nm, about 112 nm, about 113 nm, about 114 nm or about 115 nm.
12. The method of any one of claims 3 to 11, wherein the mean particle size of the lipid vesicle particles is ≤100 nm.
13. The method of any one of claims 3 to 12, wherein the lipid vesicle particles comprise a synthetic lipid, a phospholipid, or a mixture of phospholipids.
14. The method of claim 13, wherein the lipid vesicles particles comprise dioleoyl phosphatidylcholine (DOPC) and/or lecithin.
15. The method of claim 13 or 14, wherein the lipid vesicle particles further comprise cholesterol.
16. The method of claim 15, wherein the lipid vesicle particles comprise DOPC and cholesterol at a DOPC:cholesterol ratio of 10:1 (w/w).
17. The method of any one of claims 3 to 16, wherein the lipid vesicle particles of step (i) are prepared from a liposome precursor.
18. The method of claim 17, wherein the liposome precursor is Presome®.
19. The method of any one of claims 3 to 18, wherein the lipid vesicle particles are liposomes.
20. The method of claim 19, wherein the liposomes are unilamellar, multilamellar, multivesicular, or a mixture thereof.
21. The method of any one of claims 3 to 20, wherein the at least one therapeutic agent is solubilized in one or more of sodium acetate, sodium phosphate or sodium hydroxide.
22. The method of any one of claims 3 to 21, wherein the at least one therapeutic agent is solubilized in one or more of 0.1 M sodium hydroxide, 100 mM sodium acetate having a pH of 6.0 ± 1.0, 100 mM sodium acetate having a pH of 9.5 ± 1.0, 50 mM sodium phosphate having a pH of 7.0 ± 1.0 or 100 mM sodium phosphate having a pH of 6.0 ± 1.0.
23. The method of any one of claims 3 to 22, wherein the mixing of step (ii) is performed in a sodium acetate or sodium phosphate solution.
24. The method of claim 23, wherein the mixing of step (ii) is performed in 25-250 mM sodium acetate having a pH in the range of 6.0-10.5 or 25-250 mM sodium phosphate having a pH in the range of 6.0-8.0.
25. The method of claim 23, wherein the mixing of step (ii) is performed in 50 mM sodium acetate having a pH of 6.0 ± 1.0, 100 mM sodium acetate having a pH of 9.5 ± 1.0, 50 mM sodium phosphate having a pH of 7.0 ± 1.0 or 100 mM sodium phosphate having a pH of 6.0 ± 1.0.
26. The method of claim 23, wherein the mixing of step (ii) is performed in 50 mM sodium phosphate having a pH of 7.0, 100 mM sodium phosphate having a pH of 6.0, 50 mM sodium acetate having a pH of 6.0, or 100 mM sodium acetate having a pH of 9.5.
27. The method of any one of claims 3 to 26, wherein the at least one therapeutic agent comprises an antigen, a small molecule drug, an antibody or a functional fragment thereof, an antibody mimetic or a functional fragment thereof, an immunomodulatory agent, a polynucleotide encoding a polypeptide, or an interfering polynucleotide.
28. The method of any one of claims 3 to 2265, wherein the at least one therapeutic agent comprises one or more peptide antigens.
29. The method of claim 28, wherein the one or more peptide antigens are 20-30 amino acids in length.
30. The method of claim 28 or 29, wherein the one or more peptide antigens comprise neoantigens.
31. The method of claim 28, wherein the one or more peptide antigens are derived from human papillomavirus (HPV), human immunodeficiency virus (HIV), respiratory syncytial virus (RSV), bacillus anthracis, Plasmodium, or a survivin polypeptide.
32. The method of claim 31, wherein the one or more peptide antigens are FTELTLGEF, LMLGEFLKL, RISTFKNWPK, STFKNWPFL or LPPAWQPFL; or any combination thereof.
33. The method of claim 31, wherein the one or more peptide antigens are NKLCEYNVFHNKTFELPRARVNT and/or NKLSEHKTFCNKTLEQGQMYQINT.
34. The method of any one of claims 28 to 31, wherein step (ii) comprises mixing five or more different solubilized peptide antigens with the lipid vesicle particles.
35. The method of claim 34, wherein step (ii) comprises mixing up to 30 different solubilized peptide antigens with the lipid vesicle particles.
36. The method of claim 34, wherein step (ii) comprises mixing 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 different solubilized peptide antigens with the lipid vesicle particles.
37. The method of any one of claims 34 to 36, wherein, after step (ii), each of the different solubilized peptide antigens is at a concentration of at least about 0.1 mg/ml, 0.2 mg/ml, 0.3 mg/ml, 0.4 mg/ml, 0.5 mg/ml, 0.6 mg/ml, 0.7 mg/ml, 0.8 mg/ml, 0.9 mg/ml or 1.0 mg/ml.
38. The method of any one of claims 34 to 37, wherein, after step (ii), each of the different solubilized peptide antigens is at a concentration of about 0.5 mg/ml.
39. The method of any one of claims 34 to 38, wherein the different solubilized peptide antigens are not pre-selected based on any characteristic relating to isoelectric point, solubility, stability and/or immunogenicity.
40. The method of any one of claims 34 to 39, wherein the different solubilized peptide antigens have one or more different characteristics relating to isoelectric point, solubility, stability and/or immunogenicity.
41. The method of any one of claims 34 to 40, wherein the different solubilized peptide antigens have a different length, sequence, molecular weight, charge, polarity, hydrophobicity and/or hydrophilicity.
42. The method of any one of claims 28 to 41, wherein step (ii) further comprises mixing a solubilized T-helper epitope with the lipid vesicle particles, the one or more peptide antigens, and the polyI:C polynucleotide.
43. The method of any one of claims 28 to 41, wherein step (ii) comprises mixing 10-15 neoantigens and a polyI:C polynucleotide with one solubilized T-helper epitope
44. The method of claim 42 or 43, wherein the T-helper epitope comprises the amino acid sequence AQYIKANSKFIGITEL.
45. The method of claim any one of claims 42 to 44, wherein step (ii) comprises: (ii-1) providing an antigen stock comprising the one or more peptide antigens, the polyI:C polynucleotide, and the solubilized T-helper epitope; and (ii-2) mixing the antigen stock with the lipid vesicle particles to form the mixture.
46. The method of claim 45, wherein, in step (ii-1), the antigen stock is prepared in 100 mM sodium hydroxide with each solubilized peptide antigen having a concentration of about 2.0 mg/ml.
47. The method of claim 46, wherein the antigen stock is diluted 1:1 with 50 mM sodium acetate having a pH of 6.0 ± 0.5 to provide each solubilized peptide antigen at a concentration of about 1.0 mg/ml.
48. The method of any one of claims 45 to 47, wherein after the mixing in step (ii-2) and prior to drying, the pH of the mixture is adjusted to 10 ± 1.0.
49. The method of any one of claims 3 to 48 further comprising a step of sterile filtration of the mixture formed in step (ii) prior to drying.
50. The method of any one of claims 3 to 49 further comprising, between steps (ii) and (iii), a step of confirming that the lipid vesicle particles still have a mean particle size of ≤120 nm and a polydispersity index (PDI) of ≤0.1.
51. The method of any one of claims 3 to 50, wherein the drying is performed by lyophilization, spray freeze-drying, or spray drying.
52. The method of claim 51, wherein the drying is performed by lyophilization.
53. The method of claim 52, wherein the lyophilization is performed by loading one or more containers comprising the mixture of step (ii) into a bag, sealing the bag to form a sealed unit, and lyophilizing the sealed unit in a freeze-dryer.
54. The method of claim 53, wherein the bag is a sterile, autoclaved bag.
55. The method of claim 53 or 54, wherein the freeze-dryer is a benchtop freeze dryer.
56. The method of any one of claims 53 to 55, wherein the freeze-dryer contains more than one sealed unit during the lyophilization.
57. The method of claim 56, wherein each sealed unit contains a different mixture prepared by steps (i) and (ii).
58. The method of any one of claims 3 to 57, further comprising a step of evaluating the stability of the at least one solubilized therapeutic agent before and/or after the drying of step (iii).
59. The method of claim 58, wherein the stability of the therapeutic agents is evaluated by HPLC analysis.
60. The method of claim 58 or 59, wherein the therapeutic agents are peptide antigens and at least 80% of the original peptide concentration of each peptide antigen is retained in undegraded form when evaluated before drying.
61. The method of claim 60, wherein at least 75% of the original peptide concentration of each peptide antigen is retained in undegraded form when evaluated immediately after drying.
62. The method of claim 60 or 61, wherein at least 70% of the original peptide concentration of each peptide antigen is retained in undegraded form when evaluated three months after drying.
63. The method of any one of claims 60 to 62, wherein one or more of the peptide antigens shows no degradation for up to 3 months after drying.
64. The method of claim 1 or 2, wherein providing the first dried preparation comprises: (i) providing a lipid vesicle particle preparation comprising lipid vesicle particles and at least one solubilized first therapeutic agent; (ii) sizing the lipid vesicle particle preparation to form a sized lipid vesicle particle preparation comprising sized lipid vesicle particles and said at least one solubilized first therapeutic agent, said sized lipid vesicle particles having a mean particle size of ≤120 nm and a polydispersity index (PDI) of ≤0.1; (iii) mixing the sized lipid vesicle particle preparation with at least one second therapeutic agent to form a mixture, wherein said at least one second therapeutic agent is solubilized in the mixture and is different from said at least one solubilized first therapeutic agent; and (iv) drying the mixture formed in step (iii) to form a first dried preparation comprising the therapeutic agents, a lipid, and a polyI:C polynucleotide, wherein the polyI:C polynucleotide is comprised in the lipid vesicle particle preparation of step (i) or wherein the polyI:C polynucleotide is mixed with the sized lipid vesicle particle preparation of step (iii) or wherein the polyI:C polynucleotide is mixed with the mixture of step (iii).
65. The method of claim 64, wherein prior to step (ii) the lipid vesicle particles are not sized.
66. The method of claim 64 or 65, wherein, in step (i), the lipid vesicle particle preparation is in sodium acetate solution or in sodium phosphate solution.
67. The method of any one of claims 64 to 66, wherein, in step (i), the lipid vesicle particle preparation is in 25-250 mM sodium acetate having a pH in the range of 6.0-10.5 or 25-250 mM sodium phosphate having a pH in the range of 6.0-8.0.
68. The method of any one of claims 64 to 67, wherein, in step (i), the lipid vesicle particle preparation is in 50 mM sodium acetate having a pH of 6.0 ± 1.0, 100 mM sodium acetate having a pH of 9.5 ± 1.0, 50 mM sodium phosphate having a pH of 7.0 ± 1.0, or 100 mM sodium phosphate having a pH of 6.0 ± 1.0.
69. The method of any one of claims 64 to 68, wherein, in step (i), the lipid vesicle particle preparation is in 100 mM sodium acetate having a pH of 9.5 ± 0.5.
70. The method of any one of claims 64 to 69, wherein step (i) comprises: (i-1) providing a therapeutic agent stock comprising the at least one solubilized first therapeutic agent and the polyI:C polynucleotide; and (i-2) mixing the therapeutic agent stock with a lipid mixture to form the lipid vesicle preparation.
71. The method of claim 70, wherein the polyI:C polynucleotide is encapsulated in the lipid vesicle particles.
72. The method of any one of claims 64 to 71, wherein, in step (i), the at least one solubilized first therapeutic agent is encapsulated in the lipid vesicle particles.
73. The method of any one of claims 64 to 72, wherein each of the at least one first and at least one second therapeutic agents is independently selected from the group consisting of an antigen, a small molecule drug, an antibody or a functional fragment thereof, an antibody mimetic or a functional fragment thereof, an immunomodulatory agent, a polynucleotide encoding a polypeptide, or an interfering polynucleotide.
74. The method of any one of claims 64 to 73, wherein each of the at least one first and at least one second therapeutic agents comprises a peptide antigen.
75. The method of any one of claims 64 to 74, wherein, in step (i), one, two, three, four or five different solubilized first therapeutic agents are in the lipid vesicle particle preparation.
76. The method of any one of claims 64 to 75, wherein, in step (i), four different solubilized first therapeutic agents are in the lipid vesicle particle preparation.
77. The method of claim 76, wherein the four different solubilized first therapeutic agents are peptide antigens, wherein the first peptide antigen comprises the amino acid sequence
FTELTLGEF; the second peptide antigen comprises the amino acid sequence LMLGEFLKL; the third peptide antigen comprises the amino acid sequence STFKNWPFL; and the fourth peptide antigen comprises the amino acid sequence LPPAWQPFL.
78. The method of any one of claims 64 to 77, wherein, in step (iii), the sized lipid vesicle particle preparation is mixed with one, two, three, four or five different second therapeutic agents.
79. The method of any one of claims 73 to 78, wherein the at least one second therapeutic agent comprises a peptide antigen comprising the amino acid sequence RISTFKNWPK.
80. The method of any one of claims 74 to 79, wherein step (iii) further comprises mixing at least one T-helper epitope with the sized lipid vesicle particle preparation, wherein the at least one T-helper epitope is solubilized in the mixture.
81. The method of claim 80, wherein the T-helper epitope comprises the amino acid sequence AQYIKANSKFIGITEL.
82. The method of claim 80 or 81, wherein step (iii) comprises: (iii-1) providing one or more peptide antigen stocks comprising a solubilized peptide antigen, and a stock comprising the T-helper epitope; and (iii-2) mixing the stocks with the sized lipid vesicle particles to form the mixture.
83. The method of claim 82, wherein the one or more therapeutic agent stocks are prepared in mild acetic acid.
84. The method of any one of claims 64 to 83, wherein the at least one second therapeutic agent is solubilized in mild acetic acid prior to mixing with the sized lipid vesicle particle preparation in step (iii).
85. The method of any one of claims 64 to 84, wherein, in step (ii), the lipid vesicle particle preparation of step (i) is sized by high pressure homogenization, sonication and/or membrane extrusion.
86. The method of claim 85, wherein, in step (ii), the lipid vesicle particle preparation of step (i) is sized by extrusion through a 0.2 µm polycarbonate membrane followed by extrusion through a 0.1 µm polycarbonate membrane.
87. The method of claim 86, wherein the lipid vesicle particle preparation is sized by extrusion through the 0.2 µm polycarbonate membrane 20 to 40 times and extrusion through the 0.1 µm polycarbonate membrane 10 to 20 times.
88. The method of claim 85, wherein, in step (ii), the lipid vesicle particle preparation of step (i) is sized by first using high pressure homogenization to obtain sized lipid vesicle particles having a mean particle size of >120 nm and a PDI of >0.1 and then sizing by extrusion (i) 5-20 times through a 0.1 μm polycarbonate membrane or (ii) 5-20 times through a 0.08 μm polycarbonate membrane.
89. The method of any one of claims 85 to 88, wherein the membrane extrusion is performed at 1000 to 5000 psi back pressure.
90. The method of any one of claims 85 to 89, wherein the at least one solubilized first therapeutic agent is soluble at alkaline pH during high pressure membrane extrusion at about 5000 psi.
91. The method of any one of claims 64 to 90, wherein the mean particle size of the sized lipid vesicle particles is between about 80 nm and about 120 nm.
92. The method of any one of claims 64 to 91, wherein the mean particle size of the sized lipid vesicle particles is about 80 nm, about 81 nm, about 82 nm, about 83 nm, about 84 nm, about 85 nm, about 86 nm, about 87 nm, about 88 nm, about 89 nm, about 90 nm, about 91 nm, about 92 nm, about 93 nm, about 94 nm, about 95 nm, about 96 nm, about 97 nm, about 98 nm, about 99 nm, about 100 nm, about 101 nm, about 102 nm, about 103 nm, about 104 nm, about 105 nm, about 106 nm, about 107 nm, about 108 nm, about 109 nm, about 110 nm, about 111 nm, about 112 nm, about 113 nm, about 114 nm or about 115 nm.
93. The method of any one of claims 64 to 92, wherein the mean particle size of the sized lipid vesicle particles is ≤100 nm.
94. The method of any one of claims 64 to 93, wherein the lipid vesicle particles comprise a synthetic lipid, a phospholipid, or a mixture of phospholipids.
95. The method of claim 94, wherein the lipid vesicle particles comprise dioleoyl phosphatidylcholine (DOPC) and/or lecithin.
96. The method of claim 94 or 95, wherein the lipid vesicle particles further comprise cholesterol.
97. The method of claim 96, wherein the lipid vesicle particles comprise DOPC and cholesterol at a DOPC:cholesterol ratio of 10:1 (w/w).
98. The method of any one of claims 64 to 97, wherein the lipid vesicle particles are liposomes.
99. The method of claim 98, wherein the liposomes are unilamellar, multilamellar, or a mixture thereof.
100. The method of any one of claims 64 to 99 further comprising a step of sterile filtration of the mixture formed in step (iii) prior to drying.
101. The method of any one of claims 64 to 100 further comprising, between steps (iii) and (iv), a step of confirming that the sized lipid vesicle particles still have a mean particle size of ≤120 nm and a polydispersity index (PDI) of ≤0.1.
102. The method of any one of claims 64 to 101, wherein the drying is performed by lyophilization, spray freeze-drying, or spray drying.
103. The method of claim 102, wherein the drying is performed by lyophilization.
104. The method of claim 103, wherein the lyophilization is performed by loading one or more containers comprising the mixture of step (iii) into a bag, sealing the bag to form a sealed unit, and lyophilizing the sealed unit in a freeze-dryer.
105. The method of claim 104, wherein the bag is a sterile, autoclaved bag.
106. The method of claim 104 or 105, wherein the freeze-dryer is a benchtop freeze dryer.
107. The method of any one of claims 104 to 106, wherein the freeze-dryer contains more than one sealed unit during the lyophilization.
108. The method of claim 107, wherein each sealed unit contains a different mixture prepared by steps (i) to (iii).
109. The method of claim 1 or 2, wherein providing the first dried preparation comprises: (i) solubilizing at least one lipid in an organic solvent to produce a lipid solution; (ii) combining the lipid solution with at least one therapeutic agent and a polyI:C polynucleotide to produce a mixture; and
(iii) drying the mixture formed in step (ii) to form a dried preparation comprising at least one therapeutic agent, a lipid, and a polyI:C polynucleotide.
110. The method of claim 109, wherein the organic solvent comprises tertiary-butanol, optionally a 40% tertiary butanol solution.
111. The method of claim 109 or 110, wherein the at least one therapeutic agent is solubilized in DMSO or an aqueous solution prior to combining with the lipid solution.
112. The method of any one of claims 109 to 111, wherein the polyI:C polynucleotide is solubilized in water or an aqueous solution prior to combining with the lipid solution.
113. The method of any one of claims 109 to 112, wherein the at least one lipid comprises a synthetic lipid, a phospholipid, or a mixture of phospholipids.
114. The method of claim 113, wherein the at least one lipid comprises dioleoyl phosphatidylcholine (DOPC) and/or lecithin.
115. The method of claim 113 or 114, wherein the at least one lipid further comprise cholesterol.
116. The method of claim 115, wherein the at least one lipid comprises DOPC and cholesterol at a DOPC:cholesterol ratio of 10:1 (w/w).
117. The method of any one of claims 109 to 116, wherein the at least one therapeutic agent comprises an antigen, a small molecule drug, an antibody or a functional fragment thereof, an antibody mimetic or a functional fragment thereof, an immunomodulatory agent, a polynucleotide encoding a polypeptide, or an interfering polynucleotide.
118. The method of any one of claims 109 to 116, wherein the at least one therapeutic agent comprises a peptide antigen comprising the amino acid sequence AKXVAAWTLKAAARAHYNIVTF wherein ‘X’ is phenylalanine or cyclohexylalanine.
119. The method of any one of claims 109 to 118, wherein the drying is performed by lyophilization, spray freeze-drying, or spray drying.
120. The method of claim 119, wherein the drying is performed by lyophilization.
121. The method of any one of claims 1 to 120, wherein providing the second dried preparation comprises:
(i) providing lipid vesicle particles having a mean particle size of ≤120 nm and a polydispersity index (PDI) of ≤0.1; (ii) mixing the lipid vesicle particles with a lipid-based adjuvant to form a mixture; and (iii) drying the mixture formed in step (ii) to form a second dried preparation comprising a lipid-based adjuvant and a lipid.
122. The method of claim 121, wherein step (i) comprises sizing lipid vesicle particles to provide the lipid vesicle particles having a mean particle size of ≤120 nm and a PDI of ≤0.1.
123. The method of claim 122, wherein the sizing is by filter-extrusion.
124. The method of claim 122 or 123, wherein the sizing is by extrusion through one or more polycarbonate membranes, such as a 0.2 µm polycarbonate membrane, a 0.1 µm polycarbonate membrane and/or a 0.08 µm polycarbonate membrane.
125. The method of any one of claims 122 to 124, wherein the sizing is by extrusion (i) 20-40 times through a 0.2 μm polycarbonate membrane, and then 10-20 times through a 0.1 μm polycarbonate membrane; or (ii) 20-40 times through a 0.2 μm polycarbonate membrane, then 10-20 times through a 0.1 μm polycarbonate membrane, and then 10-20 times through a 0.08 μm polycarbonate membrane.
126. The method of any one of claims 122 to 125, wherein the sizing is by extrusion (i) 25 times through a 0.2 μm polycarbonate membrane, and then 10 times through a 0.1 μm polycarbonate membrane or (ii) 25 times through a 0.2 μm polycarbonate membrane, then 10 times through a 0.1 μm polycarbonate membrane, and then 15 times through a 0.08 μm polycarbonate membrane.
127. The method of claim 122, wherein the sizing is performed by first using high pressure homogenization to obtain sized lipid vesicle particles having a mean particle size of >120 nm and a PDI of >0.1 and then sizing by extrusion (i) 5-20 times through a 0.1 μm polycarbonate membrane or (ii) 5-20 times through a 0.08 μm polycarbonate membrane.
128. The method of any one of claims 121 to 127, wherein the mean particle size of the lipid vesicle particles is between about 80 nm and about 120 nm.
129. The method of any one of claims 121 to 128, wherein the mean particle size of the lipid vesicle particles is about 80 nm, about 81 nm, about 82 nm, about 83 nm, about 84 nm, about 85 nm, about 86 nm, about 87 nm, about 88 nm, about 89 nm, about 90 nm, about 91 nm, about 92
nm, about 93 nm, about 94 nm, about 95 nm, about 96 nm, about 97 nm, about 98 nm, about 99 nm, about 100 nm, about 101 nm, about 102 nm, about 103 nm, about 104 nm, about 105 nm, about 106 nm, about 107 nm, about 108 nm, about 109 nm, about 110 nm, about 111 nm, about 112 nm, about 113 nm, about 114 nm or about 115 nm.
130. The method of any one of claims 121 to 129, wherein the mean particle size of the lipid vesicle particles is ≤100 nm.
131. The method of any one of claims 121 to 130, wherein the lipid vesicle particles comprise a synthetic lipid, a phospholipid, or a mixture of phospholipids.
132. The method of claim 131, wherein the lipid vesicles particles comprise dioleoyl phosphatidylcholine (DOPC) and/or lecithin.
133. The method of claim 131 or 132, wherein the lipid vesicle particles further comprise cholesterol.
134. The method of claim 133, wherein the lipid vesicle particles comprise DOPC and cholesterol at a DOPC:cholesterol ratio of 10:1 (w/w).
135. The method of any one of claims 121 to 134, wherein the lipid vesicle particles of step (i) are prepared from a liposome precursor.
136. The method of claim 135, wherein the liposome precursor is Presome®.
137. The method of any one of claims 121 to 136, wherein the lipid vesicle particles are liposomes.
138. The method of claim 137, wherein the liposomes are unilamellar, multilamellar, multivesicular, or a mixture thereof.
139. The method of any one of claims 121 to 138, wherein the lipid-based adjuvant is solubilized in an organic solvent or an aqueous solution prior to mixing with the lipid vesicle particles.
140. The method of any one of claims 121 to 139, wherein the lipid-based adjuvant is solubilized in 0.1 M sodium hydroxide, 100 mM sodium acetate having a pH of 6.0 ± 1.0, 100 mM sodium acetate having a pH of 9.5 ± 1.0, 50 mM sodium phosphate having a pH of 7.0 ± 1.0 or 100 mM sodium phosphate having a pH of 6.0 ± 1.0 prior to mixing with the lipid vesicle particles.
141. The method of any one of claims 121 to 140, wherein the mixing of step (ii) is performed in a sodium acetate or sodium phosphate solution.
142. The method of claim 141, wherein the mixing of step (ii) is performed in 25-250 mM sodium acetate having a pH in the range of 6.0-10.5 or 25-250 mM sodium phosphate having a pH in the range of 6.0-8.0.
143. The method of claim 141, wherein the mixing of step (ii) is performed in 50 mM sodium acetate having a pH of 6.0 ± 1.0, 100 mM sodium acetate having a pH of 9.5 ± 1.0, 50 mM sodium phosphate having a pH of 7.0 ± 1.0 or 100 mM sodium phosphate having a pH of 6.0 ± 1.0.
144. The method of claim 141, wherein the mixing of step (ii) is performed in 50 mM sodium phosphate having a pH of 7.0, 100 mM sodium phosphate having a pH of 6.0, 50 mM sodium acetate having a pH of 6.0, or 100 mM sodium acetate having a pH of 9.5.
145. The method of any one of claims 121 to 144 further comprising a step of sterile filtration of the mixture formed in step (ii) prior to drying.
146. The method of any one of claims 121 to 145 further comprising, between steps (ii) and (iii), a step of confirming that the lipid vesicle particles still have a mean particle size of ≤120 nm and a polydispersity index (PDI) of ≤0.1.
147. The method of any one of claims 121 to 146, wherein the drying is performed by lyophilization, spray freeze-drying, or spray drying.
148. The method of claim 147, wherein the drying is performed by lyophilization.
149. The method of claim 148, wherein the lyophilization is performed by loading one or more containers comprising the mixture of step (ii) into a bag, sealing the bag to form a sealed unit, and lyophilizing the sealed unit in a freeze-dryer.
150. The method of claim 149, wherein the bag is a sterile, autoclaved bag.
151. The method of claim 149 or 148, wherein the freeze-dryer is a benchtop freeze dryer.
152. The method of any one of claims 149 to 151, wherein the freeze-dryer contains more than one sealed unit during the lyophilization.
153. The method of claim 152, wherein each sealed unit contains a different mixture prepared by steps (i) and (ii).
154. The method of any one of claims 1 to 120, wherein providing the second dried preparation comprises: (i) providing a lipid vesicle particle preparation comprising lipid vesicle particles and a lipid-based adjuvant; (ii) sizing the lipid vesicle particle preparation to form a sized lipid vesicle particle preparation comprising sized lipid vesicle particles and said lipid-based adjuvant, said sized lipid vesicle particles having a mean particle size of ≤120 nm and a polydispersity index (PDI) of ≤0.1; and (iii) drying the sized lipid vesicle particle preparation formed in step (ii) to form a second dried preparation comprising the lipid-based adjuvant.
155. The method of claim 154, wherein prior to step (ii) the lipid vesicle particles are not sized.
156. The method of claim 154 or 155, wherein, in step (i), the lipid vesicle particle preparation is in sodium acetate solution or in sodium phosphate solution.
157. The method of any one of claims 154 to 156, wherein, in step (i), the lipid vesicle particle preparation is in 25-250 mM sodium acetate having a pH in the range of 6.0-10.5 or 25-250 mM sodium phosphate having a pH in the range of 6.0-8.0.
158. The method of any one of claims 154 to 157, wherein, in step (i), the lipid vesicle particle preparation is in 50 mM sodium acetate having a pH of 6.0 ± 1.0, 100 mM sodium acetate having a pH of 9.5 ± 1.0, 50 mM sodium phosphate having a pH of 7.0 ± 1.0, or 100 mM sodium phosphate having a pH of 6.0 ± 1.0.
159. The method of any one of claims 154 to 158, wherein, in step (i), the lipid vesicle particle preparation is in 100 mM sodium acetate having a pH of 9.5 ± 0.5.
160. The method of any one of claims 154 to 159, wherein step (i) comprises: (i-1) providing a lipid-based adjuvant stock comprising the lipid-based adjuvant; and (i-2) mixing the lipid-based adjuvant stock with a lipid mixture to form the lipid vesicle preparation.
161. The method of any one of claims 154 to 160, wherein, in step (i), the lipid-based adjuvant is encapsulated in the lipid vesicle particles.
162. The method of any one of claims 154 to 161, wherein, in step (ii), the lipid vesicle particle preparation of step (i) is sized by high pressure homogenization, sonication and/or membrane extrusion.
163. The method of claim 162, wherein, in step (ii), the lipid vesicle particle preparation of step (i) is sized by extrusion through a 0.2 µm polycarbonate membrane followed by extrusion through a 0.1 µm polycarbonate membrane.
164. The method of claim 163, wherein the lipid vesicle particle preparation is sized by extrusion through the 0.2 µm polycarbonate membrane 20 to 40 times and extrusion through the 0.1 µm polycarbonate membrane 10 to 20 times.
165. The method of claim 162, wherein, in step (ii), the lipid vesicle particle preparation of step (i) is sized by first using high pressure homogenization to obtain sized lipid vesicle particles having a mean particle size of >120 nm and a PDI of >0.1 and then sizing by extrusion (i) 5-20 times through a 0.1 μm polycarbonate membrane or (ii) 5-20 times through a 0.08 μm polycarbonate membrane.
166. The method of any one of claims 162 to 165, wherein the membrane extrusion is performed at 1000 to 5000 psi back pressure.
167. The method of any one of claims 154 to 166, wherein the mean particle size of the sized lipid vesicle particles is between about 80 nm and about 120 nm.
168. The method of any one of claims 154 to 167, wherein the mean particle size of the sized lipid vesicle particles is about 80 nm, about 81 nm, about 82 nm, about 83 nm, about 84 nm, about 85 nm, about 86 nm, about 87 nm, about 88 nm, about 89 nm, about 90 nm, about 91 nm, about 92 nm, about 93 nm, about 94 nm, about 95 nm, about 96 nm, about 97 nm, about 98 nm, about 99 nm, about 100 nm, about 101 nm, about 102 nm, about 103 nm, about 104 nm, about 105 nm, about 106 nm, about 107 nm, about 108 nm, about 109 nm, about 110 nm, about 111 nm, about 112 nm, about 113 nm, about 114 nm or about 115 nm.
169. The method of any one of claims 154 to 168, wherein the mean particle size of the sized lipid vesicle particles is ≤100 nm.
170. The method of any one of claims 154 to 169, wherein the lipid vesicle particles comprise a synthetic lipid, a phospholipid, or a mixture of phospholipids.
171. The method of claim 170, wherein the lipid vesicle particles comprise dioleoyl phosphatidylcholine (DOPC) and/or lecithin.
172. The method of claim 170 or 171, wherein the lipid vesicle particles further comprise cholesterol.
173. The method of claim 172, wherein the lipid vesicle particles comprise DOPC and cholesterol at a DOPC:cholesterol ratio of 10:1 (w/w).
174. The method of any one of claims 154 to 173, wherein the lipid vesicle particles are liposomes.
175. The method of claim 174, wherein the liposomes are unilamellar, multilamellar, or a mixture thereof.
176. The method of any one of claims 154 to 175 further comprising a step of sterile filtration of the sized lipid vesicle particle preparationformed in step (ii) prior to drying.
177. The method of any one of claims 154 to 176 further comprising, between steps (ii) and (iii), a step of confirming that the sized lipid vesicle particles still have a mean particle size of ≤120 nm and a polydispersity index (PDI) of ≤0.1.
178. The method of any one of claims 154 to 177, wherein the drying is performed by lyophilization, spray freeze-drying, or spray drying.
179. The method of claim 178, wherein the drying is performed by lyophilization.
180. The method of claim 179, wherein the lyophilization is performed by loading one or more containers comprising the mixture of step (ii) into a bag, sealing the bag to form a sealed unit, and lyophilizing the sealed unit in a freeze-dryer.
181. The method of claim 180, wherein the bag is a sterile, autoclaved bag.
182. The method of claim 180 or 181, wherein the freeze-dryer is a benchtop freeze dryer.
183. The method of any one of claims 180 to 182, wherein the freeze-dryer contains more than one sealed unit during the lyophilization.
184. The method of claim 183, wherein each sealed unit contains a different mixture prepared by steps (i) to (ii).
185. The method of any one of claims 1 to 120, wherein providing the second dried preparation comprises: (i) solubilizing at least one lipid in an organic solvent to produce a lipid solution;
(ii) combining the lipid solution with a lipid-based adjuvant to produce a mixture; and (iii) drying the mixture formed in step (ii) to form a dried preparation comprising a lipid- based adjuvant and a lipid.
186. The method of claim 185, wherein the organic solvent comprises tertiary-butanol, optionally a 40% tertiary butanol solution.
187. The method of claim 185 or 186, wherein the lipid-based adjuvant is solubilized in an organic solvent or an aqueous solution prior to combining with the lipid solution.
188. The method of any one of claims 185 to 187, wherein the at least one lipid comprises a synthetic lipid, a phospholipid, or a mixture of phospholipids.
189. The method of claim 188, wherein the at least one lipid comprises dioleoyl phosphatidylcholine (DOPC) and/or lecithin.
190. The method of claim 188 or 189, wherein the at least one lipid further comprise cholesterol.
191. The method of claim 190, wherein the at least one lipid comprises DOPC and cholesterol at a DOPC:cholesterol ratio of 10:1 (w/w).
192. The method of any one of claims 185 to 191, wherein the drying is performed by lyophilization, spray freeze-drying, or spray drying.
193. The method of claim 192, wherein the drying is performed by lyophilization.
194. The method of any one of claims 1 to 193, wherein the polyI:C polynucleotide is RNA- based polyI:C polynucleotide or DNA-based polyI:C polynucleotide.
195. The method of any one of claims 1 to 194, wherein the polyI:C polynucleotide is double- stranded and each strand is a homopolymer of inosinic or cytidylic residues.
196. The method of any one of claims 1 to 194, wherein the polyI:C polynucleotide is double- stranded and each strand is a heteropolymer comprising both inosinic and cytidylic residues.
197. The method of any one of claims 1 to 194, wherein the polyI:C polynucleotide is a mixture comprising both homopolymeric polyI:C polynucleotides and heteropolymeric polyI:C polynucleotides.
198. The method of any one of claims 1 to 197, wherein the lipid-based adjuvant comprises one or more lipopeptide(s).
199. The method of claim 198, wherein at least one of the lipopeptides comprises palmitic acid as the lipid component.
200. The method of any one of claims 1 to 199, wherein the lipid-based adjuvant comprises dipalmitoyl-S-glyceryl-cysteine (PAM2Cys) or tripalmitoyl-S-glyceryl-cysteine (PAM3Cys).
201. The method of claim 200, wherein the lipid-based adjuvant is PAM2Cys-Ser-(Lys)4 or PAM3Cys-Ser-(Lys)4.
202. The method of claim 201, wherein the lipid-based adjuvant is PAM3Cys-Ser-(Lys)4.
203. The method of any one of claims 1 to 202, wherein the hydrophobic carrier is an oil or a mixture of oils.
204. The method of claim 203, wherein the hydrophobic carrier comprises a vegetable oil, nut oil, or mineral oil.
205. The method of claim 204, wherein the hydrophobic carrier is mineral oil or is a mannide oleate in mineral oil solution.
206. The method of claim 205, wherein the carrier is Montanide® ISA 51 VG.
207. A pharmaceutical composition prepared by the method of any one of claims 1 to 206.
208. A method of inducing an antibody and/or CTL immune response in a subject comprising administering to the subject the pharmaceutical composition of claim 207.
209. The method of claim 208, which is for treating cancer or an infectious disease.
210. Use of the pharmaceutical composition of claim 207 for inducing an antibody and/or CTL immune response in a subject.
211. The use of claim 210, which is for the treatment of cancer.
212. The use of claim 210, which is for the treatment of an infectious disease.
213. A method of delivering a therapeutic agent to a subject comprising administering to the subject the pharmaceutical composition of claim 207.
214. Use of the pharmaceutical composition of claim 207 for delivering a therapeutic agent to a subject.
215. A kit for preparing the pharmaceutical composition of claim 207, the kit comprising: a container comprising a first dried preparation prepared by the method of any one of claims 3 to 120 and 194 to 206; a container comprising second dried preparation prepared by the method of any one of claims 121 to 206; and a container comprising a hydrophobic carrier.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US202363449779P | 2023-03-03 | 2023-03-03 | |
US63/449,779 | 2023-03-03 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2024186623A1 true WO2024186623A1 (en) | 2024-09-12 |
Family
ID=90717055
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2024/018048 WO2024186623A1 (en) | 2023-03-03 | 2024-03-01 | Methods of making dried pharmaceutical compositions |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2024186623A1 (en) |
Citations (8)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2004067023A2 (en) | 2003-01-30 | 2004-08-12 | Survac Aps | Survivin-derived peptides and use thereof |
WO2006081826A2 (en) | 2005-02-04 | 2006-08-10 | Survac Aps | Survivin peptide vaccine |
US20080233143A1 (en) | 2005-02-07 | 2008-09-25 | Lipotek Pty Ltd. | Adjuvanting Material |
US20100129385A1 (en) | 2005-02-08 | 2010-05-27 | Jackson David C | Immunogenic molecules |
WO2012065997A1 (en) | 2010-11-15 | 2012-05-24 | Vib Vzw | Respiratory syncytial virus vaccine |
WO2016176761A1 (en) | 2015-05-01 | 2016-11-10 | Immunovaccine Technologies Inc., | Methods for potentiating an immune response using depot-forming and non-depot-forming vaccines |
WO2017083963A1 (en) * | 2015-11-18 | 2017-05-26 | Immunovaccine Technologies Inc. | Adjuvanting systems and water-free vaccine compositions comprising a polyi:c polynucleotide adjuvant and a lipid-based adjuvant |
WO2019178677A1 (en) * | 2018-03-20 | 2019-09-26 | Immunovaccine Technologies Inc. | Methods and compositions for targeted delivery of active agents and immunomodulatory agents to lymph nodes |
-
2024
- 2024-03-01 WO PCT/US2024/018048 patent/WO2024186623A1/en unknown
Patent Citations (9)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2004067023A2 (en) | 2003-01-30 | 2004-08-12 | Survac Aps | Survivin-derived peptides and use thereof |
WO2006081826A2 (en) | 2005-02-04 | 2006-08-10 | Survac Aps | Survivin peptide vaccine |
US20080233143A1 (en) | 2005-02-07 | 2008-09-25 | Lipotek Pty Ltd. | Adjuvanting Material |
US20110200632A1 (en) | 2005-02-07 | 2011-08-18 | Lipotek Pty Ltd. | Adjuvanting material |
US20100129385A1 (en) | 2005-02-08 | 2010-05-27 | Jackson David C | Immunogenic molecules |
WO2012065997A1 (en) | 2010-11-15 | 2012-05-24 | Vib Vzw | Respiratory syncytial virus vaccine |
WO2016176761A1 (en) | 2015-05-01 | 2016-11-10 | Immunovaccine Technologies Inc., | Methods for potentiating an immune response using depot-forming and non-depot-forming vaccines |
WO2017083963A1 (en) * | 2015-11-18 | 2017-05-26 | Immunovaccine Technologies Inc. | Adjuvanting systems and water-free vaccine compositions comprising a polyi:c polynucleotide adjuvant and a lipid-based adjuvant |
WO2019178677A1 (en) * | 2018-03-20 | 2019-09-26 | Immunovaccine Technologies Inc. | Methods and compositions for targeted delivery of active agents and immunomodulatory agents to lymph nodes |
Non-Patent Citations (2)
Title |
---|
BOWEN P: "Particle Size Distribution Measurement from Millimeters to Nanometers and from Rods to Platelets", JOURNAL OF DISPERSION SCIENCE AND TECHNOLOGY, TAYLOR AND FRANCIS GROUP, NEW YORK, NY, US, vol. 23, no. 5, 1 January 2002 (2002-01-01), pages 631 - 662, XP009102859, ISSN: 0193-2691, Retrieved from the Internet <URL:https://www.tandfonline.com/doi/full/10.1081/DIS-120015368> DOI: 10.1081/DIS-120015368 * |
CASTLE ET AL.: "Exploiting the Mutanome for Tumor Vaccination", CANCER RES, vol. 72, no. 5, 2012, pages 1081 - 1091, XP055231746, DOI: 10.1158/0008-5472.CAN-11-3722 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
JP7409594B2 (en) | Pharmaceutical compositions, methods for preparation using lipid vesicle particles of defined size, and uses thereof | |
US20210251899A1 (en) | Polymersomes comprising a soluble encapsulated antigen as well as methods of making and uses thereof | |
US20230000769A1 (en) | Oil-in-water emulsion formulations for delivery of active or therapeutic agents | |
US20200353062A1 (en) | Pharmaceutical compositions, methods for preparation comprising sizing of lipid vesicle particles, and uses thereof | |
JP2024069198A (en) | Methods and compositions for targeted delivery of active agents and immunomodulatory agents to lymph nodes | |
US20230381309A1 (en) | Methods of treating diffuse large b-cell lymphoma | |
WO2024186623A1 (en) | Methods of making dried pharmaceutical compositions | |
WO2024186646A1 (en) | Methods of making lipid adjuvanted compositions | |
Turánek et al. | Application of liposomes for construction of vaccines | |
CA3119910A1 (en) | Methods for improving the efficacy of a survivin therapeutic in the treatment of tumors | |
WO2024092352A1 (en) | Compositions that recruit and activate antigen presenting cells | |
EP4322922A1 (en) | Polymersomes comprising a soluble encapsulated polynucleotide and an ionizable lipid as well as methods of making and uses thereof | |
EP4408460A1 (en) | Survivin and mage-a9 dual-targeted immunotherapy |