WO2024209418A1 - Heating and cooling jacket for fluid vessels - Google Patents
Heating and cooling jacket for fluid vessels Download PDFInfo
- Publication number
- WO2024209418A1 WO2024209418A1 PCT/IB2024/053349 IB2024053349W WO2024209418A1 WO 2024209418 A1 WO2024209418 A1 WO 2024209418A1 IB 2024053349 W IB2024053349 W IB 2024053349W WO 2024209418 A1 WO2024209418 A1 WO 2024209418A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- jacket
- heating
- channels
- fluid
- shell
- Prior art date
Links
- 238000001816 cooling Methods 0.000 title claims abstract description 184
- 238000010438 heat treatment Methods 0.000 title claims abstract description 175
- 239000012530 fluid Substances 0.000 title claims abstract description 162
- 150000002632 lipids Chemical class 0.000 claims description 122
- 238000000034 method Methods 0.000 claims description 118
- 230000008569 process Effects 0.000 claims description 78
- 239000000203 mixture Substances 0.000 claims description 70
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N EtOH Substances CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 claims description 41
- 238000002156 mixing Methods 0.000 claims description 39
- 238000000338 in vitro Methods 0.000 claims description 35
- 238000010364 biochemical engineering Methods 0.000 claims description 34
- 238000013518 transcription Methods 0.000 claims description 34
- 230000035897 transcription Effects 0.000 claims description 34
- 238000009295 crossflow filtration Methods 0.000 claims description 29
- 230000015572 biosynthetic process Effects 0.000 claims description 22
- 239000000463 material Substances 0.000 claims description 18
- 239000000411 inducer Substances 0.000 claims description 11
- 238000011144 upstream manufacturing Methods 0.000 claims description 10
- 230000008878 coupling Effects 0.000 claims description 9
- 238000010168 coupling process Methods 0.000 claims description 9
- 238000005859 coupling reaction Methods 0.000 claims description 9
- 238000012993 chemical processing Methods 0.000 claims description 8
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 claims description 8
- 229910001220 stainless steel Inorganic materials 0.000 claims description 7
- 239000010935 stainless steel Substances 0.000 claims description 7
- 235000004443 Ricinus communis Nutrition 0.000 claims description 6
- 239000003921 oil Substances 0.000 claims description 6
- XUIMIQQOPSSXEZ-UHFFFAOYSA-N Silicon Chemical compound [Si] XUIMIQQOPSSXEZ-UHFFFAOYSA-N 0.000 claims description 5
- 229910052710 silicon Inorganic materials 0.000 claims description 5
- 239000010703 silicon Substances 0.000 claims description 5
- AEDZKIACDBYJLQ-UHFFFAOYSA-N ethane-1,2-diol;hydrate Chemical compound O.OCCO AEDZKIACDBYJLQ-UHFFFAOYSA-N 0.000 claims description 4
- 239000002861 polymer material Substances 0.000 claims description 2
- 240000000528 Ricinus communis Species 0.000 claims 2
- 229920002477 rna polymer Polymers 0.000 description 113
- 150000007523 nucleic acids Chemical class 0.000 description 58
- 108090000765 processed proteins & peptides Proteins 0.000 description 57
- 102000004196 processed proteins & peptides Human genes 0.000 description 57
- 229920001184 polypeptide Polymers 0.000 description 56
- 102000039446 nucleic acids Human genes 0.000 description 53
- 108020004707 nucleic acids Proteins 0.000 description 53
- 239000003795 chemical substances by application Substances 0.000 description 49
- 238000002360 preparation method Methods 0.000 description 48
- 210000004027 cell Anatomy 0.000 description 44
- 238000004519 manufacturing process Methods 0.000 description 43
- 108010047041 Complementarity Determining Regions Proteins 0.000 description 40
- -1 for example Chemical class 0.000 description 36
- 239000002105 nanoparticle Substances 0.000 description 35
- 239000000243 solution Substances 0.000 description 29
- 238000001914 filtration Methods 0.000 description 26
- 108020004414 DNA Proteins 0.000 description 25
- 239000000427 antigen Substances 0.000 description 25
- 108091007433 antigens Proteins 0.000 description 25
- 102000036639 antigens Human genes 0.000 description 25
- 239000000872 buffer Substances 0.000 description 25
- 230000007935 neutral effect Effects 0.000 description 25
- 229920000642 polymer Polymers 0.000 description 25
- 108090000623 proteins and genes Proteins 0.000 description 25
- 239000000523 sample Substances 0.000 description 22
- 239000007789 gas Substances 0.000 description 21
- 238000005516 engineering process Methods 0.000 description 19
- 239000002773 nucleotide Substances 0.000 description 18
- 125000003729 nucleotide group Chemical group 0.000 description 18
- 239000003814 drug Substances 0.000 description 17
- 239000000047 product Substances 0.000 description 17
- 150000001413 amino acids Chemical class 0.000 description 15
- 108020004999 messenger RNA Proteins 0.000 description 15
- 241000282414 Homo sapiens Species 0.000 description 14
- 238000012986 modification Methods 0.000 description 14
- 230000004048 modification Effects 0.000 description 13
- 239000000126 substance Substances 0.000 description 13
- 230000009467 reduction Effects 0.000 description 12
- 150000003431 steroids Chemical class 0.000 description 12
- 239000013598 vector Substances 0.000 description 12
- 108091028043 Nucleic acid sequence Proteins 0.000 description 11
- 238000009472 formulation Methods 0.000 description 11
- 239000012634 fragment Substances 0.000 description 11
- 230000014509 gene expression Effects 0.000 description 11
- 125000002652 ribonucleotide group Chemical group 0.000 description 11
- 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 description 10
- 102000004169 proteins and genes Human genes 0.000 description 10
- 239000007864 aqueous solution Substances 0.000 description 9
- 239000011148 porous material Substances 0.000 description 9
- 108091026890 Coding region Proteins 0.000 description 8
- 102000004190 Enzymes Human genes 0.000 description 8
- 108090000790 Enzymes Proteins 0.000 description 8
- 108091028664 Ribonucleotide Proteins 0.000 description 8
- 125000003275 alpha amino acid group Chemical group 0.000 description 8
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 8
- 230000006870 function Effects 0.000 description 8
- 229920001223 polyethylene glycol Polymers 0.000 description 8
- 102000040430 polynucleotide Human genes 0.000 description 8
- 108091033319 polynucleotide Proteins 0.000 description 8
- 238000012545 processing Methods 0.000 description 8
- 239000002336 ribonucleotide Substances 0.000 description 8
- 230000001225 therapeutic effect Effects 0.000 description 8
- 230000027455 binding Effects 0.000 description 7
- 230000000295 complement effect Effects 0.000 description 7
- 238000007905 drug manufacturing Methods 0.000 description 7
- 229940126534 drug product Drugs 0.000 description 7
- 229940088679 drug related substance Drugs 0.000 description 7
- 238000005259 measurement Methods 0.000 description 7
- 239000000825 pharmaceutical preparation Substances 0.000 description 7
- 239000002157 polynucleotide Substances 0.000 description 7
- 238000000746 purification Methods 0.000 description 7
- 238000003860 storage Methods 0.000 description 7
- 238000003786 synthesis reaction Methods 0.000 description 7
- 102000004163 DNA-directed RNA polymerases Human genes 0.000 description 6
- 108090000626 DNA-directed RNA polymerases Proteins 0.000 description 6
- 239000008186 active pharmaceutical agent Substances 0.000 description 6
- 238000006243 chemical reaction Methods 0.000 description 6
- 239000007979 citrate buffer Substances 0.000 description 6
- 238000004891 communication Methods 0.000 description 6
- 239000002245 particle Substances 0.000 description 6
- 239000013558 reference substance Substances 0.000 description 6
- 238000012546 transfer Methods 0.000 description 6
- 238000011282 treatment Methods 0.000 description 6
- 208000025721 COVID-19 Diseases 0.000 description 5
- 241001465754 Metazoa Species 0.000 description 5
- 108700026244 Open Reading Frames Proteins 0.000 description 5
- 239000013543 active substance Substances 0.000 description 5
- 235000012000 cholesterol Nutrition 0.000 description 5
- 201000010099 disease Diseases 0.000 description 5
- 239000002552 dosage form Substances 0.000 description 5
- 230000000694 effects Effects 0.000 description 5
- 238000011049 filling Methods 0.000 description 5
- 239000006260 foam Substances 0.000 description 5
- 239000011521 glass Substances 0.000 description 5
- UYTPUPDQBNUYGX-UHFFFAOYSA-N guanine Chemical class O=C1NC(N)=NC2=C1N=CN2 UYTPUPDQBNUYGX-UHFFFAOYSA-N 0.000 description 5
- 238000001727 in vivo Methods 0.000 description 5
- 239000012528 membrane Substances 0.000 description 5
- 210000001519 tissue Anatomy 0.000 description 5
- 239000001226 triphosphate Substances 0.000 description 5
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 4
- 208000035473 Communicable disease Diseases 0.000 description 4
- LYCAIKOWRPUZTN-UHFFFAOYSA-N Ethylene glycol Chemical compound OCCO LYCAIKOWRPUZTN-UHFFFAOYSA-N 0.000 description 4
- 240000008821 Menyanthes trifoliata Species 0.000 description 4
- 206010028980 Neoplasm Diseases 0.000 description 4
- 229940026233 Pfizer-BioNTech COVID-19 vaccine Drugs 0.000 description 4
- ISAKRJDGNUQOIC-UHFFFAOYSA-N Uracil Chemical compound O=C1C=CNC(=O)N1 ISAKRJDGNUQOIC-UHFFFAOYSA-N 0.000 description 4
- 238000010923 batch production Methods 0.000 description 4
- 150000001875 compounds Chemical class 0.000 description 4
- OPTASPLRGRRNAP-UHFFFAOYSA-N cytosine Chemical compound NC=1C=CNC(=O)N=1 OPTASPLRGRRNAP-UHFFFAOYSA-N 0.000 description 4
- 238000011026 diafiltration Methods 0.000 description 4
- 229940079593 drug Drugs 0.000 description 4
- 150000004676 glycans Chemical class 0.000 description 4
- 230000028993 immune response Effects 0.000 description 4
- 239000007788 liquid Substances 0.000 description 4
- 210000004185 liver Anatomy 0.000 description 4
- 239000002184 metal Substances 0.000 description 4
- 229910052751 metal Inorganic materials 0.000 description 4
- 239000000546 pharmaceutical excipient Substances 0.000 description 4
- 150000004713 phosphodiesters Chemical class 0.000 description 4
- 230000002265 prevention Effects 0.000 description 4
- 238000011002 quantification Methods 0.000 description 4
- 238000013519 translation Methods 0.000 description 4
- 235000011178 triphosphate Nutrition 0.000 description 4
- 238000000108 ultra-filtration Methods 0.000 description 4
- 238000011179 visual inspection Methods 0.000 description 4
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 3
- 102000040650 (ribonucleotides)n+m Human genes 0.000 description 3
- ZDTFMPXQUSBYRL-UUOKFMHZSA-N 2-Aminoadenosine Chemical compound C12=NC(N)=NC(N)=C2N=CN1[C@@H]1O[C@H](CO)[C@@H](O)[C@H]1O ZDTFMPXQUSBYRL-UUOKFMHZSA-N 0.000 description 3
- 108020005345 3' Untranslated Regions Proteins 0.000 description 3
- UUKWKUSGGZNXGA-UHFFFAOYSA-N 3,5-dinitrobenzamide Chemical compound NC(=O)C1=CC([N+]([O-])=O)=CC([N+]([O-])=O)=C1 UUKWKUSGGZNXGA-UHFFFAOYSA-N 0.000 description 3
- 108020004705 Codon Proteins 0.000 description 3
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 3
- 101000941029 Homo sapiens Endoplasmic reticulum junction formation protein lunapark Proteins 0.000 description 3
- 108060003951 Immunoglobulin Proteins 0.000 description 3
- 241000699666 Mus <mouse, genus> Species 0.000 description 3
- 102100030991 Nucleolar and spindle-associated protein 1 Human genes 0.000 description 3
- 108091034117 Oligonucleotide Proteins 0.000 description 3
- 241000283973 Oryctolagus cuniculus Species 0.000 description 3
- 108091093037 Peptide nucleic acid Proteins 0.000 description 3
- 239000004743 Polypropylene Substances 0.000 description 3
- 108010003723 Single-Domain Antibodies Proteins 0.000 description 3
- HEMHJVSKTPXQMS-UHFFFAOYSA-M Sodium hydroxide Chemical compound [OH-].[Na+] HEMHJVSKTPXQMS-UHFFFAOYSA-M 0.000 description 3
- 230000001580 bacterial effect Effects 0.000 description 3
- 201000011510 cancer Diseases 0.000 description 3
- KRKNYBCHXYNGOX-UHFFFAOYSA-N citric acid Chemical compound OC(=O)CC(O)(C(O)=O)CC(O)=O KRKNYBCHXYNGOX-UHFFFAOYSA-N 0.000 description 3
- 239000012809 cooling fluid Substances 0.000 description 3
- 208000035475 disorder Diseases 0.000 description 3
- 239000000284 extract Substances 0.000 description 3
- 230000036541 health Effects 0.000 description 3
- 102000018358 immunoglobulin Human genes 0.000 description 3
- 239000012535 impurity Substances 0.000 description 3
- 238000002955 isolation Methods 0.000 description 3
- 210000004379 membrane Anatomy 0.000 description 3
- 238000003801 milling Methods 0.000 description 3
- 229920001155 polypropylene Polymers 0.000 description 3
- 238000003908 quality control method Methods 0.000 description 3
- 238000005057 refrigeration Methods 0.000 description 3
- 239000012465 retentate Substances 0.000 description 3
- 210000002966 serum Anatomy 0.000 description 3
- 150000003384 small molecules Chemical class 0.000 description 3
- 238000011146 sterile filtration Methods 0.000 description 3
- 238000003756 stirring Methods 0.000 description 3
- 238000006467 substitution reaction Methods 0.000 description 3
- 235000000346 sugar Nutrition 0.000 description 3
- 239000000725 suspension Substances 0.000 description 3
- 229940124597 therapeutic agent Drugs 0.000 description 3
- 230000000699 topical effect Effects 0.000 description 3
- 229960005486 vaccine Drugs 0.000 description 3
- 230000003612 virological effect Effects 0.000 description 3
- NRJAVPSFFCBXDT-HUESYALOSA-N 1,2-distearoyl-sn-glycero-3-phosphocholine Chemical compound CCCCCCCCCCCCCCCCCC(=O)OC[C@H](COP([O-])(=O)OCC[N+](C)(C)C)OC(=O)CCCCCCCCCCCCCCCCC NRJAVPSFFCBXDT-HUESYALOSA-N 0.000 description 2
- ZAYHVCMSTBRABG-JXOAFFINSA-N 5-methylcytidine Chemical compound O=C1N=C(N)C(C)=CN1[C@H]1[C@H](O)[C@H](O)[C@@H](CO)O1 ZAYHVCMSTBRABG-JXOAFFINSA-N 0.000 description 2
- KDCGOANMDULRCW-UHFFFAOYSA-N 7H-purine Chemical compound N1=CNC2=NC=NC2=C1 KDCGOANMDULRCW-UHFFFAOYSA-N 0.000 description 2
- 229930024421 Adenine Natural products 0.000 description 2
- GFFGJBXGBJISGV-UHFFFAOYSA-N Adenine Chemical compound NC1=NC=NC2=C1N=CN2 GFFGJBXGBJISGV-UHFFFAOYSA-N 0.000 description 2
- 241000180579 Arca Species 0.000 description 2
- 229940022962 COVID-19 vaccine Drugs 0.000 description 2
- 108091033380 Coding strand Proteins 0.000 description 2
- 208000001528 Coronaviridae Infections Diseases 0.000 description 2
- 102000004127 Cytokines Human genes 0.000 description 2
- 108090000695 Cytokines Proteins 0.000 description 2
- 102000053602 DNA Human genes 0.000 description 2
- 241000196324 Embryophyta Species 0.000 description 2
- 241000238631 Hexapoda Species 0.000 description 2
- 238000012369 In process control Methods 0.000 description 2
- 108091026898 Leader sequence (mRNA) Proteins 0.000 description 2
- 239000002202 Polyethylene glycol Substances 0.000 description 2
- 229940022005 RNA vaccine Drugs 0.000 description 2
- 208000037847 SARS-CoV-2-infection Diseases 0.000 description 2
- 108020004459 Small interfering RNA Proteins 0.000 description 2
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 2
- 229910000831 Steel Inorganic materials 0.000 description 2
- CZMRCDWAGMRECN-UGDNZRGBSA-N Sucrose Chemical compound O[C@H]1[C@H](O)[C@@H](CO)O[C@@]1(CO)O[C@@H]1[C@H](O)[C@@H](O)[C@H](O)[C@@H](CO)O1 CZMRCDWAGMRECN-UGDNZRGBSA-N 0.000 description 2
- 229930006000 Sucrose Natural products 0.000 description 2
- 210000001744 T-lymphocyte Anatomy 0.000 description 2
- IQFYYKKMVGJFEH-XLPZGREQSA-N Thymidine Chemical compound O=C1NC(=O)C(C)=CN1[C@@H]1O[C@H](CO)[C@@H](O)C1 IQFYYKKMVGJFEH-XLPZGREQSA-N 0.000 description 2
- 229960000643 adenine Drugs 0.000 description 2
- OIRDTQYFTABQOQ-KQYNXXCUSA-N adenosine Chemical compound C1=NC=2C(N)=NC=NC=2N1[C@@H]1O[C@H](CO)[C@@H](O)[C@H]1O OIRDTQYFTABQOQ-KQYNXXCUSA-N 0.000 description 2
- 239000002671 adjuvant Substances 0.000 description 2
- 238000003556 assay Methods 0.000 description 2
- 230000031018 biological processes and functions Effects 0.000 description 2
- 238000001574 biopsy Methods 0.000 description 2
- 238000004422 calculation algorithm Methods 0.000 description 2
- 230000003197 catalytic effect Effects 0.000 description 2
- 125000002091 cationic group Chemical group 0.000 description 2
- 230000001413 cellular effect Effects 0.000 description 2
- 239000003153 chemical reaction reagent Substances 0.000 description 2
- 230000000052 comparative effect Effects 0.000 description 2
- 239000002299 complementary DNA Substances 0.000 description 2
- 238000013329 compounding Methods 0.000 description 2
- 238000007906 compression Methods 0.000 description 2
- 230000006835 compression Effects 0.000 description 2
- 230000021615 conjugation Effects 0.000 description 2
- 230000001276 controlling effect Effects 0.000 description 2
- 239000002826 coolant Substances 0.000 description 2
- 229940104302 cytosine Drugs 0.000 description 2
- 238000004925 denaturation Methods 0.000 description 2
- 230000036425 denaturation Effects 0.000 description 2
- 238000001514 detection method Methods 0.000 description 2
- 238000011161 development Methods 0.000 description 2
- 238000011143 downstream manufacturing Methods 0.000 description 2
- 238000005553 drilling Methods 0.000 description 2
- 230000007613 environmental effect Effects 0.000 description 2
- 230000002255 enzymatic effect Effects 0.000 description 2
- 239000013604 expression vector Substances 0.000 description 2
- 210000003722 extracellular fluid Anatomy 0.000 description 2
- 230000004927 fusion Effects 0.000 description 2
- 238000009396 hybridization Methods 0.000 description 2
- 210000004408 hybridoma Anatomy 0.000 description 2
- WGCNASOHLSPBMP-UHFFFAOYSA-N hydroxyacetaldehyde Natural products OCC=O WGCNASOHLSPBMP-UHFFFAOYSA-N 0.000 description 2
- 238000010965 in-process control Methods 0.000 description 2
- 238000011065 in-situ storage Methods 0.000 description 2
- 230000001965 increasing effect Effects 0.000 description 2
- 230000006698 induction Effects 0.000 description 2
- 230000001939 inductive effect Effects 0.000 description 2
- 239000011810 insulating material Substances 0.000 description 2
- 238000007918 intramuscular administration Methods 0.000 description 2
- 238000001990 intravenous administration Methods 0.000 description 2
- 238000005304 joining Methods 0.000 description 2
- 108700021021 mRNA Vaccine Proteins 0.000 description 2
- 239000007769 metal material Substances 0.000 description 2
- 210000002445 nipple Anatomy 0.000 description 2
- 229910052757 nitrogen Inorganic materials 0.000 description 2
- 238000004806 packaging method and process Methods 0.000 description 2
- 239000012466 permeate Substances 0.000 description 2
- 238000002823 phage display Methods 0.000 description 2
- 239000012071 phase Substances 0.000 description 2
- 239000002953 phosphate buffered saline Substances 0.000 description 2
- 239000013612 plasmid Substances 0.000 description 2
- 238000011084 recovery Methods 0.000 description 2
- 230000001105 regulatory effect Effects 0.000 description 2
- 230000010076 replication Effects 0.000 description 2
- 238000013341 scale-up Methods 0.000 description 2
- 238000007789 sealing Methods 0.000 description 2
- 210000003491 skin Anatomy 0.000 description 2
- 230000000087 stabilizing effect Effects 0.000 description 2
- 238000010561 standard procedure Methods 0.000 description 2
- 239000010959 steel Substances 0.000 description 2
- 238000007920 subcutaneous administration Methods 0.000 description 2
- 239000005720 sucrose Substances 0.000 description 2
- 238000012360 testing method Methods 0.000 description 2
- 238000010257 thawing Methods 0.000 description 2
- RWQNBRDOKXIBIV-UHFFFAOYSA-N thymine Chemical compound CC1=CNC(=O)NC1=O RWQNBRDOKXIBIV-UHFFFAOYSA-N 0.000 description 2
- 229940035893 uracil Drugs 0.000 description 2
- 230000000007 visual effect Effects 0.000 description 2
- 238000005406 washing Methods 0.000 description 2
- RIFDKYBNWNPCQK-IOSLPCCCSA-N (2r,3s,4r,5r)-2-(hydroxymethyl)-5-(6-imino-3-methylpurin-9-yl)oxolane-3,4-diol Chemical compound C1=2N(C)C=NC(=N)C=2N=CN1[C@@H]1O[C@H](CO)[C@@H](O)[C@H]1O RIFDKYBNWNPCQK-IOSLPCCCSA-N 0.000 description 1
- ASWBNKHCZGQVJV-UHFFFAOYSA-N (3-hexadecanoyloxy-2-hydroxypropyl) 2-(trimethylazaniumyl)ethyl phosphate Chemical compound CCCCCCCCCCCCCCCC(=O)OCC(O)COP([O-])(=O)OCC[N+](C)(C)C ASWBNKHCZGQVJV-UHFFFAOYSA-N 0.000 description 1
- RKSLVDIXBGWPIS-UAKXSSHOSA-N 1-[(2r,3r,4s,5r)-3,4-dihydroxy-5-(hydroxymethyl)oxolan-2-yl]-5-iodopyrimidine-2,4-dione Chemical compound O[C@@H]1[C@H](O)[C@@H](CO)O[C@H]1N1C(=O)NC(=O)C(I)=C1 RKSLVDIXBGWPIS-UAKXSSHOSA-N 0.000 description 1
- QLOCVMVCRJOTTM-TURQNECASA-N 1-[(2r,3r,4s,5r)-3,4-dihydroxy-5-(hydroxymethyl)oxolan-2-yl]-5-prop-1-ynylpyrimidine-2,4-dione Chemical compound O=C1NC(=O)C(C#CC)=CN1[C@H]1[C@H](O)[C@H](O)[C@@H](CO)O1 QLOCVMVCRJOTTM-TURQNECASA-N 0.000 description 1
- PISWNSOQFZRVJK-XLPZGREQSA-N 1-[(2r,4s,5r)-4-hydroxy-5-(hydroxymethyl)oxolan-2-yl]-5-methyl-2-sulfanylidenepyrimidin-4-one Chemical compound S=C1NC(=O)C(C)=CN1[C@@H]1O[C@H](CO)[C@@H](O)C1 PISWNSOQFZRVJK-XLPZGREQSA-N 0.000 description 1
- UVBYMVOUBXYSFV-XUTVFYLZSA-N 1-methylpseudouridine Chemical compound O=C1NC(=O)N(C)C=C1[C@H]1[C@H](O)[C@H](O)[C@@H](CO)O1 UVBYMVOUBXYSFV-XUTVFYLZSA-N 0.000 description 1
- UVBYMVOUBXYSFV-UHFFFAOYSA-N 1-methylpseudouridine Natural products O=C1NC(=O)N(C)C=C1C1C(O)C(O)C(CO)O1 UVBYMVOUBXYSFV-UHFFFAOYSA-N 0.000 description 1
- YKBGVTZYEHREMT-KVQBGUIXSA-N 2'-deoxyguanosine Chemical compound C1=NC=2C(=O)NC(N)=NC=2N1[C@H]1C[C@H](O)[C@@H](CO)O1 YKBGVTZYEHREMT-KVQBGUIXSA-N 0.000 description 1
- UFBJCMHMOXMLKC-UHFFFAOYSA-N 2,4-dinitrophenol Chemical compound OC1=CC=C([N+]([O-])=O)C=C1[N+]([O-])=O UFBJCMHMOXMLKC-UHFFFAOYSA-N 0.000 description 1
- JRYMOPZHXMVHTA-DAGMQNCNSA-N 2-amino-7-[(2r,3r,4s,5r)-3,4-dihydroxy-5-(hydroxymethyl)oxolan-2-yl]-1h-pyrrolo[2,3-d]pyrimidin-4-one Chemical compound C1=CC=2C(=O)NC(N)=NC=2N1[C@@H]1O[C@H](CO)[C@@H](O)[C@H]1O JRYMOPZHXMVHTA-DAGMQNCNSA-N 0.000 description 1
- RHFUOMFWUGWKKO-XVFCMESISA-N 2-thiocytidine Chemical compound S=C1N=C(N)C=CN1[C@H]1[C@H](O)[C@H](O)[C@@H](CO)O1 RHFUOMFWUGWKKO-XVFCMESISA-N 0.000 description 1
- 108700022172 2019-nCoV Vaccine mRNA-1273 Proteins 0.000 description 1
- 238000010146 3D printing Methods 0.000 description 1
- LMMLLWZHCKCFQA-UGKPPGOTSA-N 4-amino-1-[(2r,3r,4s,5r)-3,4-dihydroxy-5-(hydroxymethyl)-2-prop-1-ynyloxolan-2-yl]pyrimidin-2-one Chemical compound C1=CC(N)=NC(=O)N1[C@]1(C#CC)O[C@H](CO)[C@@H](O)[C@H]1O LMMLLWZHCKCFQA-UGKPPGOTSA-N 0.000 description 1
- XXSIICQLPUAUDF-TURQNECASA-N 4-amino-1-[(2r,3r,4s,5r)-3,4-dihydroxy-5-(hydroxymethyl)oxolan-2-yl]-5-prop-1-ynylpyrimidin-2-one Chemical compound O=C1N=C(N)C(C#CC)=CN1[C@H]1[C@H](O)[C@H](O)[C@@H](CO)O1 XXSIICQLPUAUDF-TURQNECASA-N 0.000 description 1
- CKTSBUTUHBMZGZ-ULQXZJNLSA-N 4-amino-1-[(2r,4s,5r)-4-hydroxy-5-(hydroxymethyl)oxolan-2-yl]-5-tritiopyrimidin-2-one Chemical compound O=C1N=C(N)C([3H])=CN1[C@@H]1O[C@H](CO)[C@@H](O)C1 CKTSBUTUHBMZGZ-ULQXZJNLSA-N 0.000 description 1
- 108020003589 5' Untranslated Regions Proteins 0.000 description 1
- ZAYHVCMSTBRABG-UHFFFAOYSA-N 5-Methylcytidine Natural products O=C1N=C(N)C(C)=CN1C1C(O)C(O)C(CO)O1 ZAYHVCMSTBRABG-UHFFFAOYSA-N 0.000 description 1
- AGFIRQJZCNVMCW-UAKXSSHOSA-N 5-bromouridine Chemical compound O[C@@H]1[C@H](O)[C@@H](CO)O[C@H]1N1C(=O)NC(=O)C(Br)=C1 AGFIRQJZCNVMCW-UAKXSSHOSA-N 0.000 description 1
- FHIDNBAQOFJWCA-UAKXSSHOSA-N 5-fluorouridine Chemical compound O[C@@H]1[C@H](O)[C@@H](CO)O[C@H]1N1C(=O)NC(=O)C(F)=C1 FHIDNBAQOFJWCA-UAKXSSHOSA-N 0.000 description 1
- KDOPAZIWBAHVJB-UHFFFAOYSA-N 5h-pyrrolo[3,2-d]pyrimidine Chemical compound C1=NC=C2NC=CC2=N1 KDOPAZIWBAHVJB-UHFFFAOYSA-N 0.000 description 1
- BXJHWYVXLGLDMZ-UHFFFAOYSA-N 6-O-methylguanine Chemical compound COC1=NC(N)=NC2=C1NC=N2 BXJHWYVXLGLDMZ-UHFFFAOYSA-N 0.000 description 1
- QGWBEETXHOVFQS-UHFFFAOYSA-N 6-[6-(2-hexyldecanoyloxy)hexyl-(4-hydroxybutyl)amino]hexyl 2-hexyldecanoate Chemical compound CCCCCCCCC(CCCCCC)C(=O)OCCCCCCN(CCCCO)CCCCCCOC(=O)C(CCCCCC)CCCCCCCC QGWBEETXHOVFQS-UHFFFAOYSA-N 0.000 description 1
- UEHOMUNTZPIBIL-UUOKFMHZSA-N 6-amino-9-[(2r,3r,4s,5r)-3,4-dihydroxy-5-(hydroxymethyl)oxolan-2-yl]-7h-purin-8-one Chemical compound O=C1NC=2C(N)=NC=NC=2N1[C@@H]1O[C@H](CO)[C@@H](O)[C@H]1O UEHOMUNTZPIBIL-UUOKFMHZSA-N 0.000 description 1
- OGHAROSJZRTIOK-KQYNXXCUSA-O 7-methylguanosine Chemical compound C1=2N=C(N)NC(=O)C=2[N+](C)=CN1[C@@H]1O[C@H](CO)[C@@H](O)[C@H]1O OGHAROSJZRTIOK-KQYNXXCUSA-O 0.000 description 1
- HCAJQHYUCKICQH-VPENINKCSA-N 8-Oxo-7,8-dihydro-2'-deoxyguanosine Chemical compound C1=2NC(N)=NC(=O)C=2NC(=O)N1[C@H]1C[C@H](O)[C@@H](CO)O1 HCAJQHYUCKICQH-VPENINKCSA-N 0.000 description 1
- HDZZVAMISRMYHH-UHFFFAOYSA-N 9beta-Ribofuranosyl-7-deazaadenin Natural products C1=CC=2C(N)=NC=NC=2N1C1OC(CO)C(O)C1O HDZZVAMISRMYHH-UHFFFAOYSA-N 0.000 description 1
- 241000251468 Actinopterygii Species 0.000 description 1
- GUBGYTABKSRVRQ-XLOQQCSPSA-N Alpha-Lactose Chemical compound O[C@@H]1[C@@H](O)[C@@H](O)[C@@H](CO)O[C@H]1O[C@@H]1[C@@H](CO)O[C@H](O)[C@H](O)[C@H]1O GUBGYTABKSRVRQ-XLOQQCSPSA-N 0.000 description 1
- 108091093088 Amplicon Proteins 0.000 description 1
- 108020005544 Antisense RNA Proteins 0.000 description 1
- 108091023037 Aptamer Proteins 0.000 description 1
- 241000193830 Bacillus <bacterium> Species 0.000 description 1
- 241000894006 Bacteria Species 0.000 description 1
- 241001678559 COVID-19 virus Species 0.000 description 1
- 108091033409 CRISPR Proteins 0.000 description 1
- 238000010354 CRISPR gene editing Methods 0.000 description 1
- 241000283707 Capra Species 0.000 description 1
- 108090000994 Catalytic RNA Proteins 0.000 description 1
- 102000053642 Catalytic RNA Human genes 0.000 description 1
- 241000282693 Cercopithecidae Species 0.000 description 1
- 241000251730 Chondrichthyes Species 0.000 description 1
- 241000699800 Cricetinae Species 0.000 description 1
- 150000008574 D-amino acids Chemical class 0.000 description 1
- HMFHBZSHGGEWLO-SOOFDHNKSA-N D-ribofuranose Chemical class OC[C@H]1OC(O)[C@H](O)[C@@H]1O HMFHBZSHGGEWLO-SOOFDHNKSA-N 0.000 description 1
- 108010053770 Deoxyribonucleases Proteins 0.000 description 1
- 102000016911 Deoxyribonucleases Human genes 0.000 description 1
- 239000004593 Epoxy Substances 0.000 description 1
- 241000283073 Equus caballus Species 0.000 description 1
- 241000588724 Escherichia coli Species 0.000 description 1
- 241000206602 Eukaryota Species 0.000 description 1
- 108091029865 Exogenous DNA Proteins 0.000 description 1
- 108010008177 Fd immunoglobulins Proteins 0.000 description 1
- 108010010803 Gelatin Proteins 0.000 description 1
- 108700023863 Gene Components Proteins 0.000 description 1
- WQZGKKKJIJFFOK-GASJEMHNSA-N Glucose Natural products OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 1
- 108020005004 Guide RNA Proteins 0.000 description 1
- 241000282412 Homo Species 0.000 description 1
- 108010054477 Immunoglobulin Fab Fragments Proteins 0.000 description 1
- 102000001706 Immunoglobulin Fab Fragments Human genes 0.000 description 1
- 108010021625 Immunoglobulin Fragments Proteins 0.000 description 1
- 102000008394 Immunoglobulin Fragments Human genes 0.000 description 1
- 229930010555 Inosine Natural products 0.000 description 1
- 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 1
- 108091092195 Intron Proteins 0.000 description 1
- 241000235058 Komagataella pastoris Species 0.000 description 1
- 150000008575 L-amino acids Chemical class 0.000 description 1
- GUBGYTABKSRVRQ-QKKXKWKRSA-N Lactose Natural products OC[C@H]1O[C@@H](O[C@H]2[C@H](O)[C@@H](O)C(O)O[C@@H]2CO)[C@H](O)[C@@H](O)[C@H]1O GUBGYTABKSRVRQ-QKKXKWKRSA-N 0.000 description 1
- 241000124008 Mammalia Species 0.000 description 1
- 108700011259 MicroRNAs Proteins 0.000 description 1
- 229940026207 Moderna COVID-19 vaccine Drugs 0.000 description 1
- 108091005461 Nucleic proteins Proteins 0.000 description 1
- 241001452677 Ogataea methanolica Species 0.000 description 1
- 241001494479 Pecora Species 0.000 description 1
- 108091005804 Peptidases Proteins 0.000 description 1
- 206010035226 Plasma cell myeloma Diseases 0.000 description 1
- 239000004952 Polyamide Substances 0.000 description 1
- 241000288906 Primates Species 0.000 description 1
- 239000004365 Protease Substances 0.000 description 1
- 229940096437 Protein S Drugs 0.000 description 1
- 208000008425 Protein deficiency Diseases 0.000 description 1
- CZPWVGJYEJSRLH-UHFFFAOYSA-N Pyrimidine Chemical compound C1=CN=CN=C1 CZPWVGJYEJSRLH-UHFFFAOYSA-N 0.000 description 1
- 108091034057 RNA (poly(A)) Proteins 0.000 description 1
- 108020005161 RNA Caps Proteins 0.000 description 1
- 108020004511 Recombinant DNA Proteins 0.000 description 1
- 102100037486 Reverse transcriptase/ribonuclease H Human genes 0.000 description 1
- PYMYPHUHKUWMLA-LMVFSUKVSA-N Ribose Natural products OC[C@@H](O)[C@@H](O)[C@@H](O)C=O PYMYPHUHKUWMLA-LMVFSUKVSA-N 0.000 description 1
- 108091005634 SARS-CoV-2 receptor-binding domains Proteins 0.000 description 1
- 240000004808 Saccharomyces cerevisiae Species 0.000 description 1
- 235000014680 Saccharomyces cerevisiae Nutrition 0.000 description 1
- 101000629318 Severe acute respiratory syndrome coronavirus 2 Spike glycoprotein Proteins 0.000 description 1
- 102000039471 Small Nuclear RNA Human genes 0.000 description 1
- 102000042773 Small Nucleolar RNA Human genes 0.000 description 1
- 108020003224 Small Nucleolar RNA Proteins 0.000 description 1
- 108091027967 Small hairpin RNA Proteins 0.000 description 1
- 101710198474 Spike protein Proteins 0.000 description 1
- 229920002472 Starch Polymers 0.000 description 1
- 108091081024 Start codon Proteins 0.000 description 1
- 101000677856 Stenotrophomonas maltophilia (strain K279a) Actin-binding protein Smlt3054 Proteins 0.000 description 1
- 229930182558 Sterol Natural products 0.000 description 1
- 241000187747 Streptomyces Species 0.000 description 1
- 108091008874 T cell receptors Proteins 0.000 description 1
- 102000016266 T-Cell Antigen Receptors Human genes 0.000 description 1
- 108091046869 Telomeric non-coding RNA Proteins 0.000 description 1
- 108091036066 Three prime untranslated region Proteins 0.000 description 1
- 108700009124 Transcription Initiation Site Proteins 0.000 description 1
- 241000255993 Trichoplusia ni Species 0.000 description 1
- 239000007983 Tris buffer Substances 0.000 description 1
- 206010046865 Vaccinia virus infection Diseases 0.000 description 1
- JLCPHMBAVCMARE-UHFFFAOYSA-N [3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-hydroxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methyl [5-(6-aminopurin-9-yl)-2-(hydroxymethyl)oxolan-3-yl] hydrogen phosphate Polymers Cc1cn(C2CC(OP(O)(=O)OCC3OC(CC3OP(O)(=O)OCC3OC(CC3O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c3nc(N)[nH]c4=O)C(COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3CO)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cc(C)c(=O)[nH]c3=O)n3cc(C)c(=O)[nH]c3=O)n3ccc(N)nc3=O)n3cc(C)c(=O)[nH]c3=O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)O2)c(=O)[nH]c1=O JLCPHMBAVCMARE-UHFFFAOYSA-N 0.000 description 1
- 239000006096 absorbing agent Substances 0.000 description 1
- 230000021736 acetylation Effects 0.000 description 1
- 238000006640 acetylation reaction Methods 0.000 description 1
- 230000009471 action Effects 0.000 description 1
- 239000000654 additive Substances 0.000 description 1
- 230000000996 additive effect Effects 0.000 description 1
- HMFHBZSHGGEWLO-UHFFFAOYSA-N alpha-D-Furanose-Ribose Natural products OCC1OC(O)C(O)C1O HMFHBZSHGGEWLO-UHFFFAOYSA-N 0.000 description 1
- WNROFYMDJYEPJX-UHFFFAOYSA-K aluminium hydroxide Chemical compound [OH-].[OH-].[OH-].[Al+3] WNROFYMDJYEPJX-UHFFFAOYSA-K 0.000 description 1
- 230000009435 amidation Effects 0.000 description 1
- 238000007112 amidation reaction Methods 0.000 description 1
- 230000003321 amplification Effects 0.000 description 1
- 238000004458 analytical method Methods 0.000 description 1
- 210000004102 animal cell Anatomy 0.000 description 1
- 238000000137 annealing Methods 0.000 description 1
- 230000005875 antibody response Effects 0.000 description 1
- 210000000612 antigen-presenting cell Anatomy 0.000 description 1
- PYMYPHUHKUWMLA-WDCZJNDASA-N arabinose Chemical class OC[C@@H](O)[C@@H](O)[C@H](O)C=O PYMYPHUHKUWMLA-WDCZJNDASA-N 0.000 description 1
- PYMYPHUHKUWMLA-UHFFFAOYSA-N arabinose Natural products OCC(O)C(O)C(O)C=O PYMYPHUHKUWMLA-UHFFFAOYSA-N 0.000 description 1
- QVGXLLKOCUKJST-UHFFFAOYSA-N atomic oxygen Chemical compound [O] QVGXLLKOCUKJST-UHFFFAOYSA-N 0.000 description 1
- 230000009286 beneficial effect Effects 0.000 description 1
- SRBFZHDQGSBBOR-UHFFFAOYSA-N beta-D-Pyranose-Lyxose Natural products OC1COC(O)C(O)C1O SRBFZHDQGSBBOR-UHFFFAOYSA-N 0.000 description 1
- WQZGKKKJIJFFOK-VFUOTHLCSA-N beta-D-glucose Chemical compound OC[C@H]1O[C@@H](O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-VFUOTHLCSA-N 0.000 description 1
- 230000001588 bifunctional effect Effects 0.000 description 1
- 239000013060 biological fluid Substances 0.000 description 1
- 238000005219 brazing Methods 0.000 description 1
- 238000011095 buffer preparation Methods 0.000 description 1
- 239000008364 bulk solution Substances 0.000 description 1
- 238000004364 calculation method Methods 0.000 description 1
- 238000009566 cancer vaccine Methods 0.000 description 1
- 229940022399 cancer vaccine Drugs 0.000 description 1
- 239000002775 capsule Substances 0.000 description 1
- 150000001720 carbohydrates Chemical class 0.000 description 1
- 235000014633 carbohydrates Nutrition 0.000 description 1
- 238000005266 casting Methods 0.000 description 1
- 238000004113 cell culture Methods 0.000 description 1
- 230000007969 cellular immunity Effects 0.000 description 1
- 230000036755 cellular response Effects 0.000 description 1
- 150000005829 chemical entities Chemical class 0.000 description 1
- 238000004587 chromatography analysis Methods 0.000 description 1
- 210000000349 chromosome Anatomy 0.000 description 1
- 239000003184 complementary RNA Substances 0.000 description 1
- 239000002131 composite material Substances 0.000 description 1
- 238000009833 condensation Methods 0.000 description 1
- 230000005494 condensation Effects 0.000 description 1
- 239000004020 conductor Substances 0.000 description 1
- 238000011109 contamination Methods 0.000 description 1
- 238000007796 conventional method Methods 0.000 description 1
- 230000007812 deficiency Effects 0.000 description 1
- 239000007857 degradation product Substances 0.000 description 1
- 210000005258 dental pulp stem cell Anatomy 0.000 description 1
- 230000001419 dependent effect Effects 0.000 description 1
- 230000030609 dephosphorylation Effects 0.000 description 1
- 238000006209 dephosphorylation reaction Methods 0.000 description 1
- 210000004207 dermis Anatomy 0.000 description 1
- 230000000368 destabilizing effect Effects 0.000 description 1
- 239000000032 diagnostic agent Substances 0.000 description 1
- 229940039227 diagnostic agent Drugs 0.000 description 1
- 235000015872 dietary supplement Nutrition 0.000 description 1
- 238000010790 dilution Methods 0.000 description 1
- 239000012895 dilution Substances 0.000 description 1
- 239000001177 diphosphate Substances 0.000 description 1
- 235000011180 diphosphates Nutrition 0.000 description 1
- XPPKVPWEQAFLFU-UHFFFAOYSA-N diphosphoric acid Chemical group OP(O)(=O)OP(O)(O)=O XPPKVPWEQAFLFU-UHFFFAOYSA-N 0.000 description 1
- LOKCTEFSRHRXRJ-UHFFFAOYSA-I dipotassium trisodium dihydrogen phosphate hydrogen phosphate dichloride Chemical compound P(=O)(O)(O)[O-].[K+].P(=O)(O)([O-])[O-].[Na+].[Na+].[Cl-].[K+].[Cl-].[Na+] LOKCTEFSRHRXRJ-UHFFFAOYSA-I 0.000 description 1
- 238000000375 direct analysis in real time Methods 0.000 description 1
- 238000009826 distribution Methods 0.000 description 1
- 239000013583 drug formulation Substances 0.000 description 1
- 238000001035 drying Methods 0.000 description 1
- 238000012063 dual-affinity re-targeting Methods 0.000 description 1
- 108010011867 ecallantide Proteins 0.000 description 1
- 230000002500 effect on skin Effects 0.000 description 1
- 238000009760 electrical discharge machining Methods 0.000 description 1
- 230000005611 electricity Effects 0.000 description 1
- 238000004520 electroporation Methods 0.000 description 1
- 239000000839 emulsion Substances 0.000 description 1
- 238000005538 encapsulation Methods 0.000 description 1
- 239000002158 endotoxin Substances 0.000 description 1
- 230000002708 enhancing effect Effects 0.000 description 1
- 230000006862 enzymatic digestion Effects 0.000 description 1
- 238000006911 enzymatic reaction Methods 0.000 description 1
- 210000003527 eukaryotic cell Anatomy 0.000 description 1
- 238000005187 foaming Methods 0.000 description 1
- 238000005242 forging Methods 0.000 description 1
- 238000004108 freeze drying Methods 0.000 description 1
- 238000007710 freezing Methods 0.000 description 1
- 230000008014 freezing Effects 0.000 description 1
- 239000000446 fuel Substances 0.000 description 1
- 230000002538 fungal effect Effects 0.000 description 1
- 239000000499 gel Substances 0.000 description 1
- 239000008273 gelatin Substances 0.000 description 1
- 229920000159 gelatin Polymers 0.000 description 1
- 235000019322 gelatine Nutrition 0.000 description 1
- 235000011852 gelatine desserts Nutrition 0.000 description 1
- 210000004602 germ cell Anatomy 0.000 description 1
- 239000008103 glucose Substances 0.000 description 1
- 239000003292 glue Substances 0.000 description 1
- 230000005484 gravity Effects 0.000 description 1
- 239000000383 hazardous chemical Substances 0.000 description 1
- 208000014951 hematologic disease Diseases 0.000 description 1
- 150000002402 hexoses Chemical class 0.000 description 1
- 210000005260 human cell Anatomy 0.000 description 1
- 230000008348 humoral response Effects 0.000 description 1
- 230000007062 hydrolysis Effects 0.000 description 1
- 238000006460 hydrolysis reaction Methods 0.000 description 1
- 230000002209 hydrophobic effect Effects 0.000 description 1
- 230000003053 immunization Effects 0.000 description 1
- 238000002649 immunization Methods 0.000 description 1
- 230000002163 immunogen Effects 0.000 description 1
- 230000006872 improvement Effects 0.000 description 1
- 238000000126 in silico method Methods 0.000 description 1
- 238000007373 indentation Methods 0.000 description 1
- 239000012678 infectious agent Substances 0.000 description 1
- 208000015181 infectious disease Diseases 0.000 description 1
- 238000002347 injection Methods 0.000 description 1
- 239000007924 injection Substances 0.000 description 1
- 229910052500 inorganic mineral Inorganic materials 0.000 description 1
- 229960003786 inosine Drugs 0.000 description 1
- 238000003780 insertion Methods 0.000 description 1
- 230000037431 insertion Effects 0.000 description 1
- 239000012212 insulator Substances 0.000 description 1
- 238000001361 intraarterial administration Methods 0.000 description 1
- 210000002977 intracellular fluid Anatomy 0.000 description 1
- 238000010255 intramuscular injection Methods 0.000 description 1
- 239000007927 intramuscular injection Substances 0.000 description 1
- 238000007912 intraperitoneal administration Methods 0.000 description 1
- 238000007913 intrathecal administration Methods 0.000 description 1
- 230000002601 intratumoral effect Effects 0.000 description 1
- 238000007914 intraventricular administration Methods 0.000 description 1
- VBGWSQKGUZHFPS-VGMMZINCSA-N kalbitor Chemical compound C([C@H]1C(=O)N[C@@H](CC(N)=O)C(=O)N[C@H](C(N[C@@H](CC=2C=CC=CC=2)C(=O)N[C@H](C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H]2C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC=3C=CC=CC=3)C(=O)N[C@H](C(=O)N[C@@H](CC=3C=CC(O)=CC=3)C(=O)NCC(=O)NCC(=O)N[C@H]3CSSC[C@H](NC(=O)[C@@H]4CCCN4C(=O)CNC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](C)NC(=O)[C@H](CCCCN)NC(=O)[C@H](CC=4C=CC=CC=4)NC(=O)[C@H](C)NC(=O)[C@@H](NC(=O)[C@H](CC=4C=CC=CC=4)NC(=O)[C@H](CO)NC(=O)[C@H](CC=4NC=NC=4)NC(=O)[C@H](CCSC)NC(=O)[C@H](C)NC(=O)[C@@H](N)CCC(O)=O)CSSC[C@H](NC(=O)[C@H](CCSC)NC(=O)[C@H](CCCCN)NC(=O)[C@H](CCCCN)NC(=O)[C@@H](NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CO)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](CC=4C=CC=CC=4)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CC(N)=O)NC(=O)CNC(=O)[C@H](CCC(O)=O)NC3=O)CSSC2)C(=O)N[C@@H]([C@H](C)O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC(O)=O)C(O)=O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](C)C(=O)N[C@@H](C)C(=O)N[C@@H](CC=2NC=NC=2)C(=O)N2CCC[C@H]2C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC=2C3=CC=CC=C3NC=2)C(=O)N[C@@H](CC=2C=CC=CC=2)C(=O)N1)[C@@H](C)CC)[C@H](C)O)=O)[C@@H](C)CC)C1=CC=CC=C1 VBGWSQKGUZHFPS-VGMMZINCSA-N 0.000 description 1
- 229940018902 kalbitor Drugs 0.000 description 1
- 108010045069 keyhole-limpet hemocyanin Proteins 0.000 description 1
- 210000003734 kidney Anatomy 0.000 description 1
- 238000002372 labelling Methods 0.000 description 1
- 239000008101 lactose Substances 0.000 description 1
- 238000011031 large-scale manufacturing process Methods 0.000 description 1
- 238000001638 lipofection Methods 0.000 description 1
- 239000002479 lipoplex Substances 0.000 description 1
- 239000002502 liposome Substances 0.000 description 1
- 238000011068 loading method Methods 0.000 description 1
- 230000033001 locomotion Effects 0.000 description 1
- 230000001926 lymphatic effect Effects 0.000 description 1
- 238000003754 machining Methods 0.000 description 1
- 239000011159 matrix material Substances 0.000 description 1
- 208000030159 metabolic disease Diseases 0.000 description 1
- 230000002503 metabolic effect Effects 0.000 description 1
- 230000011987 methylation Effects 0.000 description 1
- 238000007069 methylation reaction Methods 0.000 description 1
- 239000002679 microRNA Substances 0.000 description 1
- 239000011707 mineral Substances 0.000 description 1
- 238000010369 molecular cloning Methods 0.000 description 1
- 238000009126 molecular therapy Methods 0.000 description 1
- 210000004400 mucous membrane Anatomy 0.000 description 1
- 210000003205 muscle Anatomy 0.000 description 1
- 238000002703 mutagenesis Methods 0.000 description 1
- 231100000350 mutagenesis Toxicity 0.000 description 1
- 230000035772 mutation Effects 0.000 description 1
- 201000000050 myeloid neoplasm Diseases 0.000 description 1
- AEMBWNDIEFEPTH-UHFFFAOYSA-N n-tert-butyl-n-ethylnitrous amide Chemical compound CCN(N=O)C(C)(C)C AEMBWNDIEFEPTH-UHFFFAOYSA-N 0.000 description 1
- 229930014626 natural product Natural products 0.000 description 1
- 238000003199 nucleic acid amplification method Methods 0.000 description 1
- 239000002777 nucleoside Substances 0.000 description 1
- 150000003833 nucleoside derivatives Chemical class 0.000 description 1
- 238000002515 oligonucleotide synthesis Methods 0.000 description 1
- 210000000056 organ Anatomy 0.000 description 1
- 239000001301 oxygen Substances 0.000 description 1
- 229910052760 oxygen Inorganic materials 0.000 description 1
- 238000007911 parenteral administration Methods 0.000 description 1
- GJVFBWCTGUSGDD-UHFFFAOYSA-L pentamethonium bromide Chemical compound [Br-].[Br-].C[N+](C)(C)CCCCC[N+](C)(C)C GJVFBWCTGUSGDD-UHFFFAOYSA-L 0.000 description 1
- 239000000816 peptidomimetic Substances 0.000 description 1
- 230000010412 perfusion Effects 0.000 description 1
- 230000000737 periodic effect Effects 0.000 description 1
- 239000008194 pharmaceutical composition Substances 0.000 description 1
- 229940124531 pharmaceutical excipient Drugs 0.000 description 1
- 150000003904 phospholipids Chemical class 0.000 description 1
- 230000026731 phosphorylation Effects 0.000 description 1
- 238000006366 phosphorylation reaction Methods 0.000 description 1
- 230000010399 physical interaction Effects 0.000 description 1
- 230000004962 physiological condition Effects 0.000 description 1
- 230000006461 physiological response Effects 0.000 description 1
- 210000002381 plasma Anatomy 0.000 description 1
- 239000004033 plastic Substances 0.000 description 1
- 229920003023 plastic Polymers 0.000 description 1
- 229920001983 poloxamer Polymers 0.000 description 1
- 229920002647 polyamide Polymers 0.000 description 1
- 229920000447 polyanionic polymer Polymers 0.000 description 1
- 238000006116 polymerization reaction Methods 0.000 description 1
- 229920005862 polyol Polymers 0.000 description 1
- 150000003077 polyols Chemical class 0.000 description 1
- 229920001282 polysaccharide Polymers 0.000 description 1
- 239000005017 polysaccharide Substances 0.000 description 1
- 229920000915 polyvinyl chloride Polymers 0.000 description 1
- 239000004800 polyvinyl chloride Substances 0.000 description 1
- 238000012805 post-processing Methods 0.000 description 1
- 230000004481 post-translational protein modification Effects 0.000 description 1
- 238000007781 pre-processing Methods 0.000 description 1
- 210000001236 prokaryotic cell Anatomy 0.000 description 1
- QQONPFPTGQHPMA-UHFFFAOYSA-N propylene Natural products CC=C QQONPFPTGQHPMA-UHFFFAOYSA-N 0.000 description 1
- 125000004805 propylene group Chemical group [H]C([H])([H])C([H])([*:1])C([H])([H])[*:2] 0.000 description 1
- 108020003175 receptors Proteins 0.000 description 1
- 102000005962 receptors Human genes 0.000 description 1
- 238000003259 recombinant expression Methods 0.000 description 1
- 231100000205 reproductive and developmental toxicity Toxicity 0.000 description 1
- 238000012827 research and development Methods 0.000 description 1
- 230000002207 retinal effect Effects 0.000 description 1
- 238000010839 reverse transcription Methods 0.000 description 1
- 239000003161 ribonuclease inhibitor Substances 0.000 description 1
- 108020004418 ribosomal RNA Proteins 0.000 description 1
- 108091092562 ribozyme Proteins 0.000 description 1
- RHFUOMFWUGWKKO-UHFFFAOYSA-N s2C Natural products S=C1N=C(N)C=CN1C1C(O)C(O)C(CO)O1 RHFUOMFWUGWKKO-UHFFFAOYSA-N 0.000 description 1
- 150000003839 salts Chemical class 0.000 description 1
- 238000007790 scraping Methods 0.000 description 1
- 230000003248 secreting effect Effects 0.000 description 1
- 210000000717 sertoli cell Anatomy 0.000 description 1
- 230000035939 shock Effects 0.000 description 1
- 229920002545 silicone oil Polymers 0.000 description 1
- 239000004055 small Interfering RNA Substances 0.000 description 1
- 108091029842 small nuclear ribonucleic acid Proteins 0.000 description 1
- 239000011780 sodium chloride Substances 0.000 description 1
- 238000010532 solid phase synthesis reaction Methods 0.000 description 1
- 239000002904 solvent Substances 0.000 description 1
- 241000894007 species Species 0.000 description 1
- 230000009870 specific binding Effects 0.000 description 1
- 230000006641 stabilisation Effects 0.000 description 1
- 238000011105 stabilization Methods 0.000 description 1
- 239000008107 starch Substances 0.000 description 1
- 235000019698 starch Nutrition 0.000 description 1
- 230000001954 sterilising effect Effects 0.000 description 1
- 238000004659 sterilization and disinfection Methods 0.000 description 1
- 150000003432 sterols Chemical class 0.000 description 1
- 235000003702 sterols Nutrition 0.000 description 1
- 150000008163 sugars Chemical class 0.000 description 1
- 238000001356 surgical procedure Methods 0.000 description 1
- 238000001308 synthesis method Methods 0.000 description 1
- 230000002194 synthesizing effect Effects 0.000 description 1
- 238000002560 therapeutic procedure Methods 0.000 description 1
- RYYWUUFWQRZTIU-UHFFFAOYSA-K thiophosphate Chemical compound [O-]P([O-])([O-])=S RYYWUUFWQRZTIU-UHFFFAOYSA-K 0.000 description 1
- 229940113082 thymine Drugs 0.000 description 1
- 238000011200 topical administration Methods 0.000 description 1
- 230000009466 transformation Effects 0.000 description 1
- 230000009261 transgenic effect Effects 0.000 description 1
- 230000007704 transition Effects 0.000 description 1
- 125000002264 triphosphate group Chemical group [H]OP(=O)(O[H])OP(=O)(O[H])OP(=O)(O[H])O* 0.000 description 1
- LENZDBCJOHFCAS-UHFFFAOYSA-N tris Chemical compound OCC(N)(CO)CO LENZDBCJOHFCAS-UHFFFAOYSA-N 0.000 description 1
- PKIDNTKRVKSLDB-UHFFFAOYSA-K trisodium;2-hydroxypropane-1,2,3-tricarboxylate;hydrate Chemical compound O.[Na+].[Na+].[Na+].[O-]C(=O)CC(O)(CC([O-])=O)C([O-])=O PKIDNTKRVKSLDB-UHFFFAOYSA-K 0.000 description 1
- HDZZVAMISRMYHH-KCGFPETGSA-N tubercidin Chemical compound C1=CC=2C(N)=NC=NC=2N1[C@@H]1O[C@H](CO)[C@@H](O)[C@H]1O HDZZVAMISRMYHH-KCGFPETGSA-N 0.000 description 1
- 210000004881 tumor cell Anatomy 0.000 description 1
- 238000002255 vaccination Methods 0.000 description 1
- 208000007089 vaccinia Diseases 0.000 description 1
- 210000003501 vero cell Anatomy 0.000 description 1
- 108700026220 vif Genes Proteins 0.000 description 1
- 239000013603 viral vector Substances 0.000 description 1
- 238000003466 welding Methods 0.000 description 1
- 210000005253 yeast cell Anatomy 0.000 description 1
Classifications
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12M—APPARATUS FOR ENZYMOLOGY OR MICROBIOLOGY; APPARATUS FOR CULTURING MICROORGANISMS FOR PRODUCING BIOMASS, FOR GROWING CELLS OR FOR OBTAINING FERMENTATION OR METABOLIC PRODUCTS, i.e. BIOREACTORS OR FERMENTERS
- C12M23/00—Constructional details, e.g. recesses, hinges
- C12M23/02—Form or structure of the vessel
- C12M23/08—Flask, bottle or test tube
-
- B—PERFORMING OPERATIONS; TRANSPORTING
- B01—PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
- B01L—CHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
- B01L7/00—Heating or cooling apparatus; Heat insulating devices
- B01L7/04—Heat insulating devices, e.g. jackets for flasks
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12M—APPARATUS FOR ENZYMOLOGY OR MICROBIOLOGY; APPARATUS FOR CULTURING MICROORGANISMS FOR PRODUCING BIOMASS, FOR GROWING CELLS OR FOR OBTAINING FERMENTATION OR METABOLIC PRODUCTS, i.e. BIOREACTORS OR FERMENTERS
- C12M23/00—Constructional details, e.g. recesses, hinges
- C12M23/52—Mobile; Means for transporting the apparatus
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12M—APPARATUS FOR ENZYMOLOGY OR MICROBIOLOGY; APPARATUS FOR CULTURING MICROORGANISMS FOR PRODUCING BIOMASS, FOR GROWING CELLS OR FOR OBTAINING FERMENTATION OR METABOLIC PRODUCTS, i.e. BIOREACTORS OR FERMENTERS
- C12M41/00—Means for regulation, monitoring, measurement or control, e.g. flow regulation
- C12M41/12—Means for regulation, monitoring, measurement or control, e.g. flow regulation of temperature
- C12M41/18—Heat exchange systems, e.g. heat jackets or outer envelopes
- C12M41/22—Heat exchange systems, e.g. heat jackets or outer envelopes in contact with the bioreactor walls
-
- F—MECHANICAL ENGINEERING; LIGHTING; HEATING; WEAPONS; BLASTING
- F28—HEAT EXCHANGE IN GENERAL
- F28D—HEAT-EXCHANGE APPARATUS, NOT PROVIDED FOR IN ANOTHER SUBCLASS, IN WHICH THE HEAT-EXCHANGE MEDIA DO NOT COME INTO DIRECT CONTACT
- F28D1/00—Heat-exchange apparatus having stationary conduit assemblies for one heat-exchange medium only, the media being in contact with different sides of the conduit wall, in which the other heat-exchange medium is a large body of fluid, e.g. domestic or motor car radiators
- F28D1/06—Heat-exchange apparatus having stationary conduit assemblies for one heat-exchange medium only, the media being in contact with different sides of the conduit wall, in which the other heat-exchange medium is a large body of fluid, e.g. domestic or motor car radiators with the heat-exchange conduits forming part of, or being attached to, the tank containing the body of fluid
-
- B—PERFORMING OPERATIONS; TRANSPORTING
- B01—PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
- B01F—MIXING, e.g. DISSOLVING, EMULSIFYING OR DISPERSING
- B01F35/00—Accessories for mixers; Auxiliary operations or auxiliary devices; Parts or details of general application
- B01F35/90—Heating or cooling systems
- B01F35/92—Heating or cooling systems for heating the outside of the receptacle, e.g. heated jackets or burners
-
- B—PERFORMING OPERATIONS; TRANSPORTING
- B01—PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
- B01L—CHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
- B01L2300/00—Additional constructional details
- B01L2300/18—Means for temperature control
- B01L2300/1838—Means for temperature control using fluid heat transfer medium
- B01L2300/185—Means for temperature control using fluid heat transfer medium using a liquid as fluid
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12M—APPARATUS FOR ENZYMOLOGY OR MICROBIOLOGY; APPARATUS FOR CULTURING MICROORGANISMS FOR PRODUCING BIOMASS, FOR GROWING CELLS OR FOR OBTAINING FERMENTATION OR METABOLIC PRODUCTS, i.e. BIOREACTORS OR FERMENTERS
- C12M23/00—Constructional details, e.g. recesses, hinges
- C12M23/28—Constructional details, e.g. recesses, hinges disposable or single use
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12M—APPARATUS FOR ENZYMOLOGY OR MICROBIOLOGY; APPARATUS FOR CULTURING MICROORGANISMS FOR PRODUCING BIOMASS, FOR GROWING CELLS OR FOR OBTAINING FERMENTATION OR METABOLIC PRODUCTS, i.e. BIOREACTORS OR FERMENTERS
- C12M23/00—Constructional details, e.g. recesses, hinges
- C12M23/48—Holding appliances; Racks; Supports
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12M—APPARATUS FOR ENZYMOLOGY OR MICROBIOLOGY; APPARATUS FOR CULTURING MICROORGANISMS FOR PRODUCING BIOMASS, FOR GROWING CELLS OR FOR OBTAINING FERMENTATION OR METABOLIC PRODUCTS, i.e. BIOREACTORS OR FERMENTERS
- C12M27/00—Means for mixing, agitating or circulating fluids in the vessel
- C12M27/02—Stirrer or mobile mixing elements
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12M—APPARATUS FOR ENZYMOLOGY OR MICROBIOLOGY; APPARATUS FOR CULTURING MICROORGANISMS FOR PRODUCING BIOMASS, FOR GROWING CELLS OR FOR OBTAINING FERMENTATION OR METABOLIC PRODUCTS, i.e. BIOREACTORS OR FERMENTERS
- C12M41/00—Means for regulation, monitoring, measurement or control, e.g. flow regulation
- C12M41/48—Automatic or computerized control
Definitions
- the present disclosure provides technologies relating to a jacket that may be used in connection with the heating and cooling of bioreactors, and other biological and/or chemical processing vessels and containers.
- the jacket may be used in connection with single use drug production bottles.
- the jacket may be placed around a bioprocessing or chemical processing vessel, enabling the vessel to remain in situ.
- the fluid comprises at least one of water and silicon oil.
- the vessel comprises at least one of a bioprocessing vessel and a chemical processing vessel.
- the at least one fluid inlet comprises a first fluid inlet and a second fluid inlet
- the at least one fluid outlet comprises a first fluid outlet and a second fluid outlet
- the first fluid outlet being fluidly coupled to the first fluid inlet via a first fluid stream within the jacket
- the second fluid outlet being fluidly coupled to the second fluid inlet via a second fluid stream within the jacket
- the first fluid stream and the second fluid stream are fluidly disconnected within the jacket.
- the system includes at least one pump disposed fluidly upstream of the temperature handing unit; and at least one reservoir disposed fluidly upstream of the at least one pump.
- the system includes a temperature control unit (TCU) communicatively coupled to both the temperature handling unit and the at least one pump.
- TCU temperature control unit
- 11890709v1 1 Attorney Docket No.: 2013237-0929
- the system includes at least one support table for supporting the vessel and the jacket, wherein the at least one support table comprises at least one of a magnetic field generator and an inducer configured to be magnetically coupled to, and to cause rotation of, a magnetic stirrer disposed within the vessel.
- the vessel comprises a bioprocessing vessel comprising at last one of a bioreactor and a single use bottle.
- the vessel comprises at least one concavity.
- the jacket comprises at least one protrusion comprising contouring that corresponds to the at least one concavity of the vessel, the at least one protrusion protruding radially inward of a nominal inner diameter of the jacket.
- the present disclosure is directed to: a heating and cooling jacket comprising: a first semi-cylindrical shell comprising a first plurality of channels configured to allow a thermal fluid to flow therethrough, the first shell comprising a first fluid inlet and a first fluid outlet, each fluidly coupled to the first plurality of channels; and a second semi-cylindrical shell comprising a second plurality of channels configured to allow the thermal fluid to flow therethrough, the second shell comprising a second fluid inlet and a second fluid outlet, each fluidly coupled to the second plurality of channels; wherein the first and second shells form the jacket and are configured to both heat and cool, via the thermal fluid, at least one object disposed within the jacket.
- the jacket includes at least one hinge coupling the first shell to the second shell, wherein in a closed position, the jacket comprises a substantially cylindrical shape.
- each of the first and second pluralities of channels comprises a plurality of adjacent channels, each of which extends circumferentially around the respective first or second shell, and each of the channels comprises a substantially semicircular cross-section.
- each of the first shell and the second shell comprises an outer casing that defines a radially outer wall of the channels, and the outer casings are composed of at least one structural polymer material.
- each of the first and second shells are composed of stainless steel.
- each of the first and second pluralities of channels comprise channels with a uniform outer diameter; and each of the first and second pluralities of channels comprise at least one channel that is positioned at a different inner diameter than at least one other channel within the same shell.
- at least one of the first shell and the second shell comprises at least one through-bore that connects an interior of the shell to an exterior of the shell.
- the jacket includes at least one sensor access port disposed in the first shell and/or the second shell.
- the sensor access port may include a first rounded portion, a second rounded portion, and an elongated portion disposed between the first rounded portion and the second rounded portion.
- the present disclosure is directed to: a method of heating or cooling an object comprising: providing a cooling and heating jacket comprising a plurality of channels configured to allow flow of a thermal fluid therethrough; positioning at least one object to be heated or cooled within the jacket; circulating the thermal fluid through the plurality of channels such that the object is heated or cooled to a first temperature for a first period of time; and circulating the thermal fluid through the plurality of channels such that the object is heated or cooled to a second temperature for a second period of time; wherein at least one of the first temperature and the second temperature is below room temperature, and wherein at least one of first temperature and the second temperature is above room temperature.
- the jacket is configured to heat and cool the object to temperatures in a range from about 1 deg C to about 99 deg C.
- the vessel comprises at least one of: (1) one or more bioreactors during in vitro transcription, (2) one or more TFF cassettes during a first and/or a second tangential flow filtration process, (3) one or more mixing vessels containing a mix of lipids and RNA solution, and/or (4) a lipid- ethanol mixing vessel during an LNP formation process.
- the thermal fluid includes a water-glycol mixture and/or a water- ethylene-glycol mixture, the thermal fluid comprises a pH in a range from about 6.0 to about 8.5, and the density of the thermal fluid does not exceed 1kg/dm ⁇ 3.
- the thermal fluid includes a water-glycol mixture, and cooling and heating of the heating and cooling jacket is limited to a temperature range from about 2 deg C to about 35 deg C.
- the heating and cooling jacket disposed around the vessel accommodates a vessel volume of at least one of 1 L, 2 L, 5 L, 10 L, 20 L, and 50 L.
- the vessel includes a volume of at least one of 1 L, 2 L, 5 L, 10 L, 20 L, and 50 L.
- each channel of the plurality of channels includes a nominal diameter in a range from about 8 mm to about 20 mm.
- the heating and cooling jacket includes a plurality of vertical spaces vertically separating each channel of the plurality of channels from a channel immediately above and/or below each channel.
- each vertical space of the plurality of vertical spaces includes a height of from about 7 mm to about 10 mm.
- the heating and cooling jacket includes a substantially cylindrical shape; each channel of the plurality of channels extends circumferentially within the heating and cooling jacket 11890709v1 3 Attorney Docket No.: 2013237-0929 around at least a portion of the circumference of the heating and cooling jacket; and each channel of the plurality of channels is disposed above and/or below one or more adjacent channels of the plurality of channels.
- the system and/or jacket includes at least one continuous fluid path through the heating and cooling jacket, the continuous fluid path including each channel of the plurality of channels, wherein the plurality of channels comprises from about 5 to about 20 channels.
- the at least one continuous fluid path includes a plurality of connections disposed at circumferential end(s) of each channel of the plurality of channels, wherein each connection of the plurality of connections connects a channel of the plurality of channels to at least one other channel (i.e., an adjacent channel) of the plurality of channels.
- each connection of the plurality of connections causes the continuous fluid path to make a 180-degree turn.
- the heating and cooling jacket includes a first semi-cylindrical shell and a second semi-cylindrical shell
- the continuous fluid path includes at least two continuous fluid paths disposed through the heating and cooling jacket
- the two continuous fluid paths include a first continuous fluid path disposed through the first semi-cylindrical shell and a second continuous fluid path disposed through the second semi-cylindrical shell.
- the heating and cooling jacket includes a height in a range from about 100 mm to about 600 mm.
- the heating and cooling jacket includes a height in a range from about 230 mm to about 300 mm.
- the heating and cooling jacket includes an outer diameter in a range from about 100 mm to about 500 mm. [0041] In some embodiments, the heating and cooling jacket includes an outer diameter in a range from about 230 mm to about 330 mm. [0042] In some embodiments, the heating and cooling jacket includes a height in a range from about 290 mm to about 390 mm. [0043] In some embodiments, each channel of the plurality of channels includes a nominal diameter in a range from about 12 mm to about 15 mm. [0044] In some embodiments, the heating and cooling jacket includes material forming the walls of each channel disposed within each vertical space of the plurality of vertical spaces.
- the heating and cooling jacket includes a channel diameter to channel spacing ratio in a range from about 1.2 to about 2.5.
- the heating and cooling jacket includes a channel diameter to channel spacing ratio in a range from about 1.4 to about 1.9. 11890709v1 4 Attorney Docket No.: 2013237-0929 [0047]
- the present disclosure is directed to a mobile bioreactor system including: a movable cart; a bioreactor disposed on the moveable cart; and the jacket according to the present disclosure, where the jacket is disposed around the bioreactor.
- the jacket includes a first shell hingedly connected to a second shell, and one of the first shell and the second shell is rigidly coupled to a top surface of the movable cart.
- the movable cart includes wheels and castors that are removably coupled to the cart.
- the bioreactor includes a nominal volume of 50 L.
- the movable cart includes a vertically oriented frame for supporting a bracket, a filter holder, a support plate, and/or an electrical switchbox.
- the jacket comprises channels with an aspect ratio (height to width ratio) of greater than 2.0.
- RNA-LNP compositions for treatment and/or prevention of coronavirus infection, e.g., SARS-CoV-2 infection, as described in Walsh et al. “RNA-based COVID-19 vaccine BNT162b2 selected for a pivotal efficacy study” medRxiv preprint (2020), which is online accessible at: https://doi.org/10.1101/2020.08.17.20176651; and Milligan et al.
- Figure 1 depicts a schematic of an exemplary drug manufacturing process.
- Figure 2 depicts an overview of an automated drug manufacturing process, according to aspects of the present disclosure.
- Figure 3A illustrates a perspective view of single use bottles in a heating and cooling jacket, according to aspects of the present disclosure.
- Figure 3B illustrates a perspective view of single use bottles in a heating and cooling jacket, according to aspects of the present disclosure.
- Figure 4 illustrates a support table, according to aspects of the present disclosure.
- Figure 5A illustrates details of a single use bottle in a heating and cooling jacket, according to aspects of the present disclosure.
- Figure 5B illustrates interior details of a heating and cooling jacket, according to aspects of the present disclosure.
- 11890709v1 5 Attorney Docket No.: 2013237-0929
- Figure 6A illustrates exterior details of a heating and cooling jacket, according to aspects of the present disclosure.
- Figure 6B illustrates interior details of a heating and cooling jacket, according to aspects of the present disclosure.
- Figure 7A illustrates further details of a single use bottle in a heating and cooling jacket, according to aspects of the present disclosure.
- Figure 7B illustrates a side perspective view of a heating and cooling jacket, according to aspects of the present disclosure.
- Figure 7C illustrates details of a heating and cooling jacket sensor access port, according to aspects of the present disclosure.
- Figure 8A illustrates an image of exterior details of a heating and cooling jacket, according to aspects of the present disclosure.
- Figure 8B illustrates an image of exterior details of a heating and cooling jacket, according to aspects of the present disclosure.
- Figure 9A illustrates a perspective view of single use bottle in a heating and cooling jacket, according to aspects of the present disclosure.
- Figure 9B illustrates a perspective view of single use bottle in a heating and cooling jacket, according to aspects of the present disclosure.
- Figure 10 illustrates a system for heating and cooling single use bottles, according to aspects of the present disclosure.
- Figure 11 depicts an overview of an exemplary manufacturing process for a pharmaceutical- grade composition comprising RNA, according to aspects of the present disclosure.
- Figure 12 illustrates an overview of exemplary DNA template manufacture process via a PCR-based process, according to aspects of the present disclosure.
- Figure 13 illustrates an exemplary process for manufacturing LNP compositions, according to aspects of the present disclosure.
- Figure 14 illustrates a method or process for heating or cooling a chemical or biological process, according to aspects of the present disclosure.
- Figure 15 illustrates a heating and cooling jacket in an open position, according to aspects of the present disclosure.
- Figure 16 illustrates a heating and cooling jacket in a closed position, according to aspects of the present disclosure.
- Figure 17 illustrates a heating and cooling jacket in operation, according to aspects of the present disclosure.
- 11890709v1 6 Attorney Docket No.: 2013237-0929
- Figure 18 illustrates a heating and cooling jacket in a closed position without outer casings, according to aspects of the present disclosure.
- Figure 19 illustrates an example of a 1 L heating and cooling jacket, according to aspects of the present disclosure.
- Figure 20 illustrates an example of a 5 L heating and cooling jacket, according to aspects of the present disclosure.
- Figure 21 illustrates an example of a 10 L heating and cooling jacket, according to aspects of the present disclosure.
- Figure 22 illustrates an example of a 20 L heating and cooling jacket, according to aspects of the present disclosure.
- Figure 23 illustrates an example of grip features on a 1 L single use bottle in a heating and cooling jacket, according to aspects of the present disclosure.
- Figure 24 illustrates an example of grip concavities in a heating and cooling jacket, according to aspects of the present disclosure.
- Figure 25 illustrates an interior view of a 50 L heating and cooling jacket, according to aspects of the present disclosure.
- Figure 26 illustrates a cross-sectional view of a 50 L heating and cooling jacket, according to aspects of the present disclosure.
- Figure 27 illustrates a perspective view of a 50 L heating and cooling jacket, according to aspects of the present disclosure.
- Figure 28 illustrates a front view of a mobile bioreactor unit with a heating and cooling jacket, according to aspects of the present disclosure.
- Figure 29 illustrates a side view of a mobile bioreactor unit with a heating and cooling jacket, according to aspects of the present disclosure.
- Figure 30 illustrates a rear view of a mobile bioreactor unit with a heating and cooling jacket, according to aspects of the present disclosure.
- Figure 31 illustrates a front view of a mobile bioreactor unit with a heating and cooling jacket, according to aspects of the present disclosure.
- Figure 32 illustrates a perspective view of a mobile bioreactor unit without a heating and cooling jacket, according to aspects of the present disclosure.
- Figure 33 illustrates a partial front view of a mobile bioreactor unit with a heating and cooling jacket on load cells, according to aspects of the present disclosure.
- Figure 34 illustrates a partial front view of a mobile bioreactor unit with a heating and cooling jacket on a scale, according to aspects of the present disclosure.
- 11890709v1 7 Attorney Docket No.: 2013237-0929 Certain Definitions [0095] About or Approximately: The term “about” or “approximately”, when used herein in reference to a value, refers to a value that is similar, in context to a stated reference value.
- the term “about” or “approximately” may encompass a range of values that are within 25%, 20%, 19%, 18%, 17%, 16%, 15%, 14%, 13%, 12%, 11%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, or less of the referred value.
- Administration typically refers to the administration of a composition to a subject or system.
- administration may be ocular, oral, parenteral, topical, etc.
- administration may be bronchial (e.g., by bronchial instillation), buccal, dermal (which may be or comprise, for example, one or more of topical to the dermis, intradermal, intradermal, transdermal, etc.), enteral, intra-arterial, intradermal, intragastric, intramedullary, intramuscular, intranasal, intraperitoneal, intrathecal, intravenous, intraventricular, within a specific organ (e.
- administration may be intramuscular.
- administration may involve dosing that is intermittent (e.g., a plurality of doses separated in time) and/or periodic (e.g., individual doses separated by a common period of time) dosing.
- administration may involve continuous dosing (e.g., perfusion) for at least a selected period of time.
- agent in general, is used to refer to an entity (e.g., for example, a lipid, metal, nucleic acid, polypeptide, polysaccharide, small molecule, etc., or complex, combination, mixture or system [e.g., cell, tissue, organism] thereof), or phenomenon (e.g., heat, electric current or field, magnetic force or field, etc.).
- entity e.g., for example, a lipid, metal, nucleic acid, polypeptide, polysaccharide, small molecule, etc., or complex, combination, mixture or system [e.g., cell, tissue, organism] thereof
- phenomenon e.g., heat, electric current or field, magnetic force or field, etc.
- the term may be used to refer to a natural product in that it is found in and/or is obtained from nature.
- the term may be used to refer to one or more entities that is man-made in that it is designed, engineered, and/or produced through action of the hand of man and/or is not found in nature.
- an agent may be utilized in isolated or pure form; in some embodiments, an agent may be utilized in crude form.
- potential agents may be provided as collections or libraries, for example that may be screened to identify or characterize active agents within them.
- the term “agent” may refer to a compound or entity that is or comprises a polymer; in some cases, the term may refer to a compound or entity that comprises one or more polymeric moieties. In some embodiments, the term “agent” may refer to a compound or entity that is not a polymer and/or is substantially free of any polymer and/or of one or more particular polymeric moieties. In some embodiments, the term may refer to a compound or entity that lacks or is substantially free of any polymeric moiety. [0098] Analog: As used herein, the term “analog” refers to a substance that shares one or more particular structural features, elements, components, or moieties with a reference substance.
- an 11890709v1 8 Attorney Docket No.: 2013237-0929 “analog” shows significant structural similarity with the reference substance, for example sharing a core or consensus structure, but also differs in certain discrete ways.
- an analog is a substance that can be generated from the reference substance, e.g., by chemical manipulation of the reference substance.
- an analog is a substance that can be generated through performance of a synthetic process substantially similar to (e.g., sharing a plurality of steps with) one that generates the reference substance.
- an analog is or can be generated through performance of a synthetic process different from that used to generate the reference substance.
- Antibody agent refers to an agent that specifically binds to a particular antigen.
- the term encompasses any polypeptide or polypeptide complex that includes immunoglobulin structural elements sufficient to confer specific binding.
- Exemplary antibody agents include, but are not limited to monoclonal antibodies or polyclonal antibodies.
- an antibody agent may include one or more constant region sequences that are characteristic of mouse, rabbit, primate, or human antibodies.
- an antibody agent may include one or more sequence elements are humanized, primatized, chimeric, etc., as is known in the art.
- an antibody agent utilized in accordance with the present disclosure is in a format selected from, but not limited to, intact IgA, IgG, IgE or IgM antibodies; bi- or multi- specific antibodies (e.g., Zybodies®, etc.); antibody fragments such as Fab fragments, Fab' fragments, F(ab')2 fragments, Fd' fragments, Fd fragments, and isolated complementarity determining regions (CDRs) or sets thereof; single chain Fvs; polypeptide-Fc fusions; single domain antibodies (e.g., shark single domain antibodies such as IgNAR or fragments thereof); cameloid antibodies; masked antibodies (e.g., Probodies®); Small Modular ImmunoPharmaceuticals (“SMIPsTM”); single chain or Tandem diabodies (TandAb
- an antibody may lack a covalent modification (e.g., attachment of a glycan) that it would have if produced naturally.
- an antibody may contain a covalent modification (e.g., attachment of a glycan, a payload [e.g., a detectable moiety, a therapeutic moiety, a catalytic moiety, etc.], or other pendant group [e.g., poly-ethylene glycol, etc.].
- an antibody agent is or comprises a polypeptide whose amino acid sequence includes one or more structural elements recognized by those skilled in the art as a complementarity determining region (CDR); in some embodiments an antibody agent is or comprises a polypeptide whose amino acid sequence includes at least one CDR (e.g., at least one heavy chain CDR and/or at least one light chain CDR) that is substantially identical to one found in a reference antibody. In some embodiments an included CDR is substantially identical to a reference CDR in that it is either identical in sequence or contains between 1-5 amino acid substitutions as compared with the reference CDR.
- CDR complementarity determining region
- an included CDR is substantially identical to a reference CDR in that it shows at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity with the reference CDR. In some embodiments, an included CDR is substantially identical to a reference CDR in that it shows at least 95%, 96%, 97%, 98%, 99%, or 100% sequence identity with the reference CDR.
- an included CDR is substantially identical to a reference CDR in that at least one amino acid within the included CDR is deleted, added, or substituted as compared with the reference CDR but the included CDR has an amino acid sequence that is otherwise identical 11890709v1 9 Attorney Docket No.: 2013237-0929 with that of the reference CDR.
- an included CDR is substantially identical to a reference CDR in that 1-5 amino acids within the included CDR are deleted, added, or substituted as compared with the reference CDR but the included CDR has an amino acid sequence that is otherwise identical to the reference CDR.
- an included CDR is substantially identical to a reference CDR in that at least one amino acid within the included CDR is substituted as compared with the reference CDR but the included CDR has an amino acid sequence that is otherwise identical with that of the reference CDR.
- an included CDR is substantially identical to a reference CDR in that 1-5 amino acids within the included CDR are deleted, added, or substituted as compared with the reference CDR but the included CDR has an amino acid sequence that is otherwise identical to the reference CDR.
- an antibody agent is or comprises a polypeptide whose amino acid sequence includes structural elements recognized by those skilled in the art as an immunoglobulin variable domain.
- an antibody agent is a polypeptide protein having a binding domain which is homologous or largely homologous to an immunoglobulin-binding domain.
- Antibody agents can be made by the skilled person using methods and commercially available services and kits known in the art. For example, methods of preparation of monoclonal antibodies are well known in the art and include hybridoma technology and phage display technology. Further antibodies suitable for use in the present disclosure are described, for example, in the following publications: Antibodies A Laboratory Manual, Second edition. Edward A. Greenfield. Cold Spring Harbor Laboratory Press (September 30, 2013); Making and Using Antibodies: A Practical Handbook, Second Edition. Eds. Gary C. Howard and Matthew R. Kaser.
- Antibodies may be produced by standard techniques, for example by immunization with the appropriate polypeptide or portion(s) thereof, or by using a phage display library.
- a selected mammal e.g., mouse, rabbit, goat, horse, etc.
- various adjuvants may be used to increase immunological response.
- adjuvants include, but are not limited to, Freund's, mineral gels such as aluminum hydroxide, and surface-active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanin, and dinitrophenol. Serum from the immunized animal is collected and treated according to known procedures.
- Antigen refers to (i) an agent that elicits an immune response; and/or (ii) an agent that binds to a T cell receptor (e.g., when presented by an MHC molecule) or to an antibody.
- an antigen elicits a humoral response (e.g., including production of antigen-specific antibodies); in some embodiments, an antigen elicits a cellular response (e.g., involving T-cells whose receptors specifically interact with the antigen). In some embodiments, an antigen binds to an antibody and may or may not induce a particular physiological response in an organism.
- a humoral response e.g., including production of antigen-specific antibodies
- an antigen elicits a cellular response (e.g., involving T-cells whose receptors specifically interact with the antigen).
- an antigen binds to an antibody and may or may not induce a particular physiological response in an organism.
- an antigen may be 11890709v1 10 Attorney Docket No.: 2013237-0929 or include any chemical entity such as, for example, a small molecule, a nucleic acid, a polypeptide, a carbohydrate, a lipid, a polymer (in some embodiments other than a biologic polymer [e.g., other than a nucleic acid or amino acid polymer]) etc.
- an antigen is or comprises a polypeptide.
- an antigen is or comprises a glycan.
- an antigen may be provided in isolated or pure form, or alternatively may be provided in crude form (e.g., together with other materials, for example in an extract such as a cellular extract or other relatively crude preparation of an antigen-containing source).
- antigens utilized in accordance with the present invention are provided in a crude form.
- an antigen is a recombinant antigen.
- Bioreactor refers to a vessel used for in vitro transcription described herein.
- a bioreactor can be of any size so long as it is useful for in vitro transcription.
- a bioreactor can be at least 0.5 liter, including 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 liters or more, or any volume in between.
- Cap refers to a structure comprising or essentially consisting of a nucleoside-5 '-triphosphate that is typically joined to a 5'-end of an uncapped RNA (e.g., an uncapped RNA having a 5'- diphosphate).
- a cap is or comprises a guanine nucleotide.
- a cap is or comprises a naturally-occurring RNA 5’ cap, including, e.g., but not limited to a N7- methylguanosine cap, which has a structure designated as "m7G.”
- a cap is or comprises a synthetic cap analog that resembles an RNA cap structure and possesses the ability to stabilize RNA if attached thereto, including, e.g., but not limited to anti-reverse cap analogs (ARCAs) known in the art.
- ARCAs anti-reverse cap analogs
- a capped RNA may be obtained by in vitro capping of RNA that has a 5' triphosphate group or RNA that has a 5' diphosphate group with a capping enzyme system (including, e.g., but not limited to vaccinia capping enzyme system or Saccharomyces cerevisiae capping enzyme system).
- a capped RNA can be obtained by in vitro transcription (IVT) of a DNA template, wherein, in addition to the GTP, an IVT system also contains a cap analog, e.g., as known in the art.
- Non-limiting examples of a cap analog include a m7GpppG cap analog or an N7-methyl-, 2’-O- methyl -GpppG ARCA cap analog or an N7-methyl-, 3'-O-methyl-GpppG ARCA cap analog, or any commercially available cap analogs, including, e.g., CleanCap (Trilink), EZ Cap, etc.
- a cap analog is or comprises a trinucleotide cap analog.
- Comparable refers to two or more agents, entities, situations, sets of conditions, etc., that may not be identical to one another but that are sufficiently similar to permit comparison there between so that one skilled in the art will appreciate that conclusions may reasonably be drawn based on differences or similarities observed.
- comparable sets of conditions, circumstances, individuals, or populations are characterized by a plurality of substantially identical features and one or a small number of varied features.
- Complementary As used herein, the term “complementary” is used in reference to oligonucleotide hybridization related by base-pairing rules. For example, the sequence “C-A-G-T” is complementary to the sequence “G-T-C-A.” Complementarity can be partial or total.
- any degree of partial complementarity is intended to be included within the scope of the term “complementary” provided that the partial complementarity permits oligonucleotide hybridization.
- Partial complementarity is where one or more nucleic acid bases is not matched according to the base pairing rules.
- Total or complete complementarity between nucleic acids is where each and every nucleic acid base is matched with another base under the base pairing rules.
- Detecting The term “detecting” is used broadly herein to include appropriate means of determining the presence or absence of an entity of interest or any form of measurement of an entity of interest in a sample. Thus, “detecting” may include determining, measuring, assessing, or assaying the presence or absence, level, amount, and/or location of an entity of interest.
- Quantitative and qualitative determinations, measurements or assessments are included, including semi-quantitative. Such determinations, measurements or assessments may be relative, for example when an entity of interest is being detected relative to a control reference, or absolute.
- the term “quantifying” when used in the context of quantifying an entity of interest can refer to absolute or to relative quantification. Absolute quantification may be accomplished by correlating a detected level of an entity of interest to known control standards (e.g., through generation of a standard curve). Alternatively, relative quantification can be accomplished by comparison of detected levels or amounts between two or more different entities of interest to provide a relative quantification of each of the two or more different entities of interest, i.e., relative to each other.
- determining involves manipulation of a physical sample.
- determining involves consideration and/or manipulation of data or information, for example utilizing a computer or other processing unit adapted to perform a relevant analysis.
- determining involves receiving relevant information and/or materials from a source.
- determining involves comparing one or more features of a sample or entity to a comparable reference.
- Dosage form or unit dosage form may be used to refer to a physically discrete unit of an active agent (e.g., a therapeutic or diagnostic agent) for administration to a subject.
- an active agent e.g., a therapeutic or diagnostic agent
- each such unit contains a predetermined quantity of active agent.
- such quantity is a unit dosage amount (or a whole fraction thereof) appropriate for administration in accordance with a dosing regimen that has been determined to correlate with a desired or beneficial outcome when administered to a relevant population (i.e., with a therapeutic dosing regimen).
- Encapsulate The term “encapsulate” or “encapsulation” is used herein to refer to at least a portion of a component is enclosed or surrounded by another material or another component in a composition. In some embodiments, a component can be fully enclosed or surrounded by another material or another component in a composition.
- Excipient refers to a non-therapeutic agent that may be included in a pharmaceutical composition, for example to provide or contribute to a desired property or effect (e.g., desired consistency, delivery, and/or stabilizing effect, etc.).
- suitable pharmaceutical excipients to be added to a LNP composition may include, for example, salts, starch, glucose, lactose, sucrose, gelatin, sodium chloride, glycerol, propylene, glycol, water, ethanol and the like.
- homolog refers to the overall relatedness between polynucleotide molecules (e.g., DNA molecules and/or RNA molecules) and/or between polypeptide molecules.
- polynucleotide molecules e.g., DNA molecules and/or RNA molecules
- polypeptide molecules are considered to be “homologous” to one another if their sequences are at least 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 99% identical.
- Identity refers to the overall relatedness between polymeric molecules, e.g., between nucleic acid molecules (e.g., DNA molecules and/or RNA molecules) and/or between polypeptide molecules.
- polymeric molecules are considered to be “substantially identical” to one another if their sequences are at least 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 99% identical.
- the percent identity between the two sequences is a function of the number of identical positions shared by the sequences, taking into account the number of gaps, and the length of each gap, which needs to be introduced for optimal alignment of the two sequences.
- the comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm. For example, the percent identity between two nucleotide sequences can be determined using the algorithm of Meyers and Miller (CABIOS, 1989, 4: 11-17), which has been incorporated into the ALIGN program (version 2.0).
- nucleic acid sequence comparisons made with the ALIGN program use a PAM120 weight residue table, a gap length penalty of 12 and a gap penalty of 4.
- the percent identity between two nucleotide sequences can, alternatively, be determined using the GAP program in the GCG software package using an NWSgapdna.CMP matrix.
- these terms, or grammatically comparable comparative terms indicate values that are relative to a comparable reference measurement. For example, in some embodiments, an assessed value achieved with an agent of interest may be “improved” relative to that obtained with a comparable reference agent.
- an assessed value achieved in a subject or system of interest may be “improved” relative to that obtained in the same subject or system under different conditions (e.g., prior to or after an event such as administration of an agent of interest), or in a different, comparable subject (e.g., in a comparable subject or system that differs from the subject or system of interest in presence of one or more indicators of a particular disease, disorder or condition of interest, or in prior exposure to a condition or agent, etc.).
- comparative terms refer to statistically relevant differences (e.g., that are of a prevalence and/or magnitude sufficient to achieve statistical relevance).
- in vitro refers to events that occur in an artificial environment, e.g., in a test tube or reaction vessel (e.g., a bioreactor), in cell culture, etc., rather than within a multi-cellular organism.
- in vitro transcription refers to the process whereby transcription occurs in vitro in a non-cellular system to produce a synthetic RNA product for use in various applications, including, e.g., production of protein or polypeptides.
- synthetic RNA products can be translated in vitro or introduced directly into cells, where they can be translated.
- Such synthetic RNA products include, e.g., but not limited to mRNAs, antisense RNA molecules, shRNA molecules, long non- coding RNA molecules, ribozymes, aptamers, guide RNAs (e.g., for CRISPR), ribosomal RNAs, small nuclear RNAs, small nucleolar RNAs, and the like.
- An IVT reaction typically utilizes a DNA template (e.g., a linear DNA template) as described and/or utilized herein, ribonucleotides (e.g., non-modified ribonucleotide triphosphates or modified ribonucleotide triphosphates), and an appropriate RNA polymerase.
- in vitro transcription RNA composition refers to a composition comprising target RNA synthesized by in vitro transcription.
- a composition can comprise excess in vitro transcription reagents (including, e.g., ribonucleotides and/or capping agents), nucleic acids or fragments thereof such as DNA templates or fragments thereof, polypeptides or fragments thereof such as recombinant enzymes or host cell proteins or fragments thereof, and/or other impurities.
- an in vitro transcription RNA composition may have been treated and/or processed prior to a purification process that ultimately produces an RNA transcript preparation comprising RNA transcript at a desired concentration in an appropriate buffer for formulation and/or further manufacturing and/or processing.
- an in vitro transcription RNA composition may have been treated to remove or digest DNA template (e.g., using a DNase).
- an in vitro transcription RNA composition may have been treated to remove or digest polypeptides (e.g., enzymes such as RNA polymerases, RNase inhibitors, etc.) present in an in vitro transcription reaction (e.g., using a protease).
- Nanoparticle refers to a particle having a diameter of less than 1000 nanometers (nm). In some embodiments, a nanoparticle has a diameter of less than 300 nm, as defined by the National Science Foundation. In some embodiments, a nanoparticle has a diameter of less than 100 nm as defined by the National Institutes of Health. In some embodiments, a nanoparticle has a diameter of less than 80 nm as defined by the National Institutes of Health.
- a nanoparticle comprises one or more enclosed compartments, separated from the bulk solution by a membrane, which surrounds and encloses a space or compartment.
- Nucleic acid/ Polynucleotide refers to a polymer of at least 2 nucleotides or more, including, e.g., at least 3 nucleotides, at least 4 nucleotides, at least 5 nucleotides, at least 6 nucleotides, at least 7 nucleotides, at least 8 nucleotides, at least 9 nucleotides, at least 11890709v1 16 Attorney Docket No.: 2013237-0929 10 nucleotides, or more.
- a nucleic acid is or comprises DNA. In some embodiments, a nucleic acid is or comprises RNA. In some embodiments, a nucleic acid is or comprises peptide nucleic acid (PNA). In some embodiments, a nucleic acid is or comprises a single stranded nucleic acid. In some embodiments, a nucleic acid is or comprises a double-stranded nucleic acid. In some embodiments, a nucleic acid comprises both single and double-stranded portions. In some embodiments, a nucleic acid comprises a backbone that comprises one or more phosphodiester linkages. In some embodiments, a nucleic acid comprises a backbone that comprises both phosphodiester and non-phosphodiester linkages.
- a nucleic acid may comprise a backbone that comprises one or more phosphorothioate or 5'-N-phosphoramidite linkages and/or one or more peptide bonds, e.g., as in a “peptide nucleic acid”.
- a nucleic acid comprises one or more, or all, natural residues (e.g., adenine, cytosine, deoxyadenosine, deoxycytidine, deoxyguanosine, deoxythymidine, guanine, thymine, uracil).
- a nucleic acid comprises on or more, or all, non-natural residues.
- a non- natural residue comprises a nucleoside analog (e.g., 2-aminoadenosine, 2-thiothymidine, inosine, pyrrolo- pyrimidine, 3 -methyl adenosine, 5-methylcytidine, C-5 propynyl-cytidine, 1-methyl-pseudouridine, C-5 propynyl-uridine, 2-aminoadenosine, C5-bromouridine, C5-fluorouridine, C5-iodouridine, C5-propynyl-uridine, C5 -propynyl-cytidine, C5-methylcytidine, 2-aminoadenosine, 7-deazaadenosine, 7-deazaguanosine, 8- oxoadenosine, 8-oxoguanosine, 6-O-methylguanine, 2-thiocytidine, methylated bases, inter
- a non-natural residue comprises one or more modified sugars (e.g., 2'-fluororibose, ribose, 2'-deoxyribose, arabinose, and hexose) as compared to those in natural residues.
- a nucleic acid has a nucleotide sequence that encodes a functional gene product such as an RNA or polypeptide.
- a nucleic acid has a nucleotide sequence that comprises one or more introns.
- a nucleic acid may be prepared by isolation from a natural source, enzymatic synthesis (e.g., by polymerization based on a complementary template, e.g., in vivo or in vitro, reproduction in a recombinant cell or system, or chemical synthesis.
- enzymatic synthesis e.g., by polymerization based on a complementary template, e.g., in vivo or in vitro, reproduction in a recombinant cell or system, or chemical synthesis.
- a nucleic acid is at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 225, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475, 500, 600, 700, 800, 900, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 5500, 6000, 6500, 7000, 7500, 8000, 8500, 9000, 9500, 10,000, 10,500, 11,000, 11,500, 12,000, 12,500, 13,000, 13,500, 14,000, 14,500, 15,000, 15,500, 16,000, 16,500, 17,000, 17,500, 18,000, 18,500, 19,000, 19,500, or 20,000 or more residues or nucleotides long.
- composition grade refers to standards for chemical and biological drug substances, drug products, dosage forms, compounded preparations, excipients, medical devices, and dietary supplements, established by a recognized national or regional pharmacopeia (e.g., The United States Pharmacopeia and The Formulary (USP–NF)).
- Polypeptide The term “polypeptide”, as used herein, typically has its art-recognized meaning of a polymer of at least three amino acids or more.
- polypeptide is intended to be sufficiently general as to encompass not only polypeptides having a complete sequence recited herein, but also to encompass polypeptides that represent functional, biologically active, or characteristic fragments, portions or domains (e.g., fragments, portions, or domains retaining at least one activity) of such complete polypeptides.
- polypeptides may contain L-amino acids, D-amino acids, or both and/or may contain any of a variety of amino acid modifications or analogs known in the 11890709v1 17 Attorney Docket No.: 2013237-0929 art.
- polypeptides may comprise natural amino acids, non-natural amino acids, synthetic amino acids, and combinations thereof (e.g., may be or comprise peptidomimetics).
- a polypeptide may be or comprise an enzyme.
- a polypeptide may be or comprise a polypeptide antigen.
- a polypeptide may be or comprise an antibody agent.
- a polypeptide may be or comprise a cytokine.
- a preparation that contains more than about 90% of a particular agent or entity is typically considered to be a pure preparation.
- an agent or entity is at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% pure in a preparation.
- Ribonucleotide As used herein, the term “ribonucleotide” encompasses unmodified ribonucleotides and modified ribonucleotides.
- unmodified ribonucleotides include the purine bases adenine (A) and guanine (G), and the pyrimidine bases cytosine (C) and uracil (U).
- Modified ribonucleotides may include one or more modifications including, but not limited to, for example, (a) end modifications, e.g., 5' end modifications (e.g., phosphorylation, dephosphorylation, conjugation, inverted linkages, etc.), 3' end modifications (e.g., conjugation, inverted linkages, etc.), (b) base modifications, e.g.
- RNA Ribonucleic acid
- RNA Ribonucleic acid
- an RNA is double stranded. In some embodiments, an RNA comprises both single and double stranded portions. In some embodiments, an RNA can comprise a backbone structure as described in the definition of “Nucleic acid / Polynucleotide” above.
- An RNA can be a regulatory RNA (e.g., siRNA, microRNA, etc.), or a messenger RNA (mRNA). In some embodiments, an RNA is a mRNA. In some embodiments, where an RNA is a mRNA, an RNA typically comprises at its 3’ end a poly(A) region.
- an RNA typically comprises at its 5’ end, an art-recognized cap structure, e.g., for recognizing and attachment of a mRNA to a ribosome to initiate translation.
- an RNA is a synthetic RNA. Synthetic RNAs include RNAs that are synthesized in vitro (e.g., by enzymatic synthesis methods and/or by chemical synthesis methods).
- an RNA is a single-stranded RNA.
- a single-stranded RNA may comprise self-complementary elements and/or may establish a secondary and/or tertiary structure.
- a single-stranded RNA when referred to as “encoding,” it can mean that it comprises a nucleic acid sequence that itself encodes or that it comprises a complement of the nucleic acid sequence that encodes.
- a single-stranded RNA can be a self-amplifying RNA (also known as self-replicating RNA).
- Recombinant as used herein, is intended to refer to polypeptides that are designed, engineered, prepared, expressed, created, manufactured, and/or or isolated by recombinant means, such as 11890709v1 18 Attorney Docket No.: 2013237-0929 polypeptides expressed using a recombinant expression vector transfected into a host cell; polypeptides isolated from a recombinant, combinatorial human polypeptide library; polypeptides isolated from an animal (e.g., a mouse, rabbit, sheep, fish, etc.) that is transgenic for or otherwise has been manipulated to express a gene or genes, or gene components that encode and/or direct expression of the polypeptide or one or more component(s), portion(s), element(s), or domain(s) thereof; and/or polypeptides prepared, expressed, created or isolated by any other means that involves splicing or ligating selected nucleic acid sequence elements to one another, chemically synthesizing selected sequence elements, and/
- one or more of such selected sequence elements is found in nature. In some embodiments, one or more of such selected sequence elements is designed in silico. In some embodiments, one or more such selected sequence elements results from mutagenesis (e.g., in vivo or in vitro) of a known sequence element, e.g., from a natural or synthetic source such as, for example, in the germline of a source organism of interest (e.g., of a human, a mouse, etc.).
- mutagenesis e.g., in vivo or in vitro
- a known sequence element e.g., from a natural or synthetic source such as, for example, in the germline of a source organism of interest (e.g., of a human, a mouse, etc.).
- an agent, animal, individual, population, sample, sequence or value of interest is compared with a reference or control agent, animal, individual, population, sample, sequence or value.
- a reference or control is tested and/or determined substantially simultaneously with the testing or determination of interest.
- a reference or control is a historical reference or control, optionally embodied in a tangible medium.
- a reference or control is determined or characterized under comparable conditions or circumstances to those under assessment.
- RNA polymerase refers to an enzyme that catalyzes polyribonucleotide synthesis by addition of ribonucleotide units to a nucleotide chain using DNA or RNA as a template.
- the term refers to either a complete enzyme as it occurs in nature, or an isolated, active catalytic or functional domain, or fragment thereof.
- an RNA polymerase enzyme initiates synthesis at the 3'-end of a primer or a nucleic acid strand, or at a promoter sequence, and proceeds in the 5'- direction along the target nucleic acid to synthesize a strand complementary to the target nucleic acid until synthesis terminates.
- RNA transcript preparation refers to a preparation comprising RNA transcript that is purified from an in vitro transcription RNA composition described herein.
- an RNA transcript preparation is a preparation comprising pharmaceutical-grade RNA transcript.
- an RNA transcript preparation is a preparation comprising RNA transcript, which includes one or more product quality attributes that are characterized and determined to meet a release and/or acceptance criteria (e.g., as described herein).
- room temperature As used herein, the term “room temperature” refers to an ambient temperature.
- a room temperature is about 18 deg C-30 deg C, e.g., about 18 deg C -25 deg C, or about 20 deg C -25 deg C, or about 20 deg C -30 deg C, or about 23 deg C -27 deg C or about 25 deg C.
- Sample typically refers to an aliquot of material obtained or derived from a source of interest, e.g., as described herein.
- a source of interest is a biological or environmental source.
- a source of interest may be or comprise a cell or an organism, such as a microbe, a plant, or an animal (e.g., a mouse).
- a source of interest is or comprises biological tissue or fluid.
- a biological fluid may be or comprise an intracellular fluid, an extracellular fluid, an intravascular fluid (blood plasma), an interstitial fluid, a lymphatic fluid, and/or a transcellular fluid.
- a biological tissue or sample may be obtained, for example, by aspirate, biopsy (e.g., fine needle or tissue biopsy), swab (e.g., oral, nasal, skin, or vaginal swab), scraping, surgery, washing or lavage (e.g., bronchioalveolar, ductal, nasal, ocular, oral, uterine, vaginal, or other washing or lavage).
- a sample is or comprises cells obtained from a subject.
- a sample is a “primary sample” obtained directly from a source of interest by any appropriate means.
- sample refers to a preparation that is obtained by processing (e.g., by removing one or more components of and/or by adding one or more agents to) a primary sample.
- a “processed sample” may comprise, for example nucleic acids or proteins extracted from a sample or obtained by subjecting a primary sample to one or more techniques such as amplification or reverse transcription of nucleic acid, isolation and/or purification of certain components, etc.
- Stable when applied to nucleic acids and/or compositions comprising nucleic acids, e.g., encapsulated in lipid nanoparticles, means that such nucleic acids and/or compositions maintain one or more aspects of their characteristics (e.g., physical and/or structural characteristics, function, and/or activity) over a period of time under a designated set of conditions (e.g., pH, temperature, light, relative humidity, etc.).
- such stability is maintained over a period of time of at least about one hour; in some embodiments, such stability is maintained over a period of time of about 5 hours, about 10 hours, about one (1) day, about one (1) week, about two (2) weeks, about one (1) month, about two (2) months, about three (3) months, about four (4) months, about five (5) months, about six (6) months, about eight (8) months, about ten (10) months, about twelve (12) months, about twenty-four (24) months, about thirty-six (36) months, or longer. In some embodiments, such stability is maintained over a period of time within the range of about one (1) day to about twenty-four (24) months, about two (2) weeks to about twelve (12) months, about two (2) months to about five (5) months, etc.
- such stability is maintained under an ambient condition (e.g., at room temperature and ambient pressure). In some embodiments, such stability is maintained under a physiological condition (e.g., in vivo or at about 37 oC for example in serum or in phosphate buffered saline). In some embodiments, such stability is maintained under cold storage (e.g., at or below about 4 oC, including, e.g., -20 oC, or -70 oC). In some embodiments, such stability is maintained when nucleic acids and/or compositions comprising the same are protected from light (e.g., maintaining in the dark).
- the term “stable” is used in reference to a nanoparticle composition (e.g., a lipid nanoparticle composition).
- a stable nanoparticle 11890709v1 20 Attorney Docket No.: 2013237-0929 composition e.g., a stable nanoparticle composition
- maintain one or more aspects of its characteristics e.g., physical and/or structural characteristics, function(s), and/or activity
- characteristics e.g., physical and/or structural characteristics, function(s), and/or activity
- a stable nanoparticle composition e.g., a lipid nanoparticle composition
- average particle size, particle size distribution, and/or polydispersity of nanoparticles is substantially maintained (e.g., within 10% or less, as compared to the initial characteristic(s)) over a period of time (e.g., as described herein) under a designated set of conditions (e.g., as described herein).
- a stable nanoparticle composition e.g., a lipid nanoparticle composition
- a stable nanoparticle composition is characterized in that no detectable amount of degradation products (e.g., associated with hydrolysis and/or enzymatic digestion) is present after it is maintained under a designated set of conditions (e.g., as described herein) over a period of time.
- degradation products e.g., associated with hydrolysis and/or enzymatic digestion
- a synthetic nucleic acid or polynucleotide refers to a nucleic acid molecule that is chemically synthesized, e.g., in some embodiments by solid-phase synthesis.
- the term “synthetic” refers to an entity that is made outside of biological cells.
- a synthetic nucleic acid or polynucleotide refers to a nucleic acid molecule (e.g., an RNA) that is produced by in vitro transcription using a template.
- Tangential flow filtration As used herein, the term “tangential flow filtration (TFF)” refers to a mode of operation of a filtration system where a fluid passes parallel to a filter membrane (i.e., the flow is tangential to the membrane) to reduce clogging of the filter. In some embodiments, particles larger than the pore size will flow past the membrane as retentate and is recycled back to the feed. In some embodiments, particles smaller than the pore size will pass through and be collected as permeate.
- Three prime untranslated region refers to the sequence of an mRNA molecule that begins following the stop codon of the coding region of an open reading frame sequence. In some embodiments, the 3' UTR begins immediately after the stop codon of the coding region of an open reading frame sequence. In other embodiments, the 3' UTR does not begin immediately after stop codon of the coding region of an open reading frame sequence.
- Threshold level refers to a level that are used as a reference to attain information on and/or classify the results of a measurement, for example, the results of a measurement attained in an assay.
- a threshold level means a value measured in an assay that defines the dividing line between two subsets of a population (e.g., a batch that satisfy quality control criteria vs. a batch that does not satisfy quality control criteria).
- a value that is equal to or higher than the threshold level defines one subset of the population, and a value that is lower than the threshold level defines the other subset of the population.
- vectors are capable of autonomous replication in a host cell into which they are introduced (e.g., bacterial vectors having a bacterial origin of replication and episomal mammalian vectors).
- Other vectors e.g., non-episomal mammalian vectors
- certain vectors are capable of directing the expression of genes to which they are operatively linked. Such vectors are referred to herein as "expression vectors.”
- Standard techniques may be used for recombinant DNA, oligonucleotide synthesis, and tissue culture and transformation (e.g., electroporation, lipofection).
- Enzymatic reactions and purification techniques may be performed according to manufacturer's specifications or as commonly accomplished in the art or as described herein.
- the foregoing techniques and procedures may be generally performed according to conventional methods well known in the art and as described in various general and more specific references that are cited and discussed throughout the present specification. See e.g., Green and Sambrook, Molecular Cloning: A Laboratory Manual (4th ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (2012)), which is incorporated herein by reference for any purpose.
- RNA therapeutics represent a particularly promising class of therapies for treatment and prevention of various diseases such as cancer, infectious diseases, and/or diseases or disorders associated with overabundance or deficiency in certain proteins.
- RNA therapeutics in particular provide remarkably effective as vaccines to address the COVID19 pandemic. Particularly given the promise of this technology, and its adaptability to a wide variety of clinical contexts, including massively large scale (e.g., vaccination and/or treatment on a global scale such as is under development for SARS-CoV-2), improvements to manufacturing technologies, especially those applicable to large-scale production, are especially valuable.
- lipid nanoparticle technologies have proven to be particularly effective (reviewed in, for example, Cullis et al. Molecular Therapy 25:1467, July 5, 2017; See also, US Patent 8058069), specifically including for RNA therapeutics (reviewed in, for example, Hou et al., Nat. Rev. Mater doi.org/10.1038/s41578- 021-00358-0, August 10, 2021).
- Technologies provided herein are useful, among other things, to achieve particularly effective and/or efficient production, e.g., on commercial scale and/or under commercial conditions, of pharmaceutical grade LNP preparations and/or compositions (e.g., nucleic acid-LNP preparations, and specifically RNA-LNP preparations).
- pharmaceutical grade LNP preparations and/or compositions e.g., nucleic acid-LNP preparations, and specifically RNA-LNP preparations.
- provided technologies permit and/or facilitate achievement of requirements unique to pharmaceutical-grade (and/or scale) production such as, for example, batch size and/or rate of production, pre-determined in-process controls and/or lot release specifications (e.g., high purity, integrity, potency, and/or stability, etc.), etc.
- LNP compositions e.g., including RNA, e.g., therapeutic RNA such as therapeutic mRNA.
- provided technologies are useful for manufacturing pharmaceutical-grade RNA-LNP therapeutics.
- provided technologies are useful for large scale manufacturing of LNP (e.g., nucleic acid-LNP, e.g., RNA-LNP) therapeutics, e.g., pharmaceutical-grade therapeutics.
- technologies provided herein can be used to produce a pharmaceutical-grade batch throughput of at least 10,000 vials of LNP (e.g., nucleic acid-LNP, e.g., RNA-LNP) therapeutics (including, e.g., at least 20,000 vials, at least 30,000 vials, at least 40,000 vials, at least 50,000 vials, at least 60,000 vials, at least 70,000 vials, at least 80,000 vials, at least 90,000 vials, at least 100,000 vials, at least 200,000 vials, at least 300,000 vials, at least 400,000 vials, at least 500,000 vials, or more).
- LNP e.g., nucleic acid-LNP, e.g., RNA-LNP
- therapeutics including, e.g., at least 20,000 vials, at least 30,000 vials, at least 40,000 vials, at least 50,000 vials, at least 60,000 vials, at
- technologies provided herein can be used to produce a pharmaceutical-grade batch throughput of at least 50 L of LNP (e.g., nucleic acid-LNP, e.g., RNA-LNP) therapeutics (including e.g., at least 50 L, at least 60 L, at least 70 L, at least 80 L, at least 100 L, at least 110 L, at least 120 L, at least 130 L, at least 140 L, at least 150 L or more.
- LNP e.g., nucleic acid-LNP, e.g., RNA-LNP
- therapeutics including e.g., at least 50 L, at least 60 L, at least 70 L, at least 80 L, at least 100 L, at least 110 L, at least 120 L, at least 130 L, at least 140 L, at least 150 L or more.
- each vial can comprise an RNA drug product in an amount of 0.01 mg to 0.5 mg (e.g., 0.01 mg, 0.02 mg, 0.03 mg, 0.04 mg, 0.05 mg, 0.06 mg, 0.07 mg, 0.08 mg, 0.09 mg, 0.1 mg, 0.15 mg, 0.2 mg, 0.25 mg, 0.3 mg, 0.35 mg, 0.4 mg, 0.45 mg, 0.5 mg).
- LNP e.g., nucleic acid-LNP, e.g., RNA-LNP
- compositions for treatment and/or prevention of a disease, disorder, or condition (e.g., cancer, infectious diseases, diseases associated with protein deficiency, etc.).
- technologies described herein can be useful for manufacturing LNP (e.g., nucleic acid-LNP, e.g., RNA-LNP) compositions that comprise or deliver (e.g., by comprising and/or delivering a nucleic acid, such as an RNA, that encodes it) a polypeptide.
- LNP e.g., nucleic acid-LNP, e.g., RNA-LNP
- technologies described herein can be useful for manufacturing LNP (e.g., nucleic acid-LNP, e.g., RNA-LNP) compositions for inducing an immune response to an antigen.
- LNP nucleic acid-LNP
- RNA-LNP nucleic acid-LNP
- technologies described herein can be useful for manufacturing LNP(e.g., nucleic acid-LNP, e.g., RNA-LNP) compositions for treatment and/or prevention of coronavirus infection, e.g., SARS-CoV-2 infection, as described in Walsh et al. “RNA-based COVID-19 vaccine BNT162b2 selected for a pivotal efficacy study” medRxiv preprint (2020), which is online accessible at: https://doi.org/10.1101/2020.08.17.20176651; and Milligan et al.
- lipid nanoparticles have achieved successful clinical delivery of a wide range of therapeutic agents including, for example, small molecules, and various nucleic acids – e.g., oligonucleotides, siRNAs, and mRNAs (reviewed, for example, in Hu et al., Nat. Rev. Mater.
- LNPs are 11890709v1 23 Attorney Docket No.: 2013237-0929 parenterally administered; most clinical studies have utilized parenteral administration, and particularly intravenous, subcutaneous, intradermal, intravitreal, intratumoral, or intramuscular injection. Intrautero injection has also been described. In some embodiments, topical administration is utilized.
- intranasal administration is utilized.
- administered LNPs are delivered to or accumulate in the liver.
- liver delivery can prove useful for achieving delivery of an LNP-encapsulated agent (and/or, in the case of a nucleic acid agent such as an RNA agent, a polypeptide encoded thereby) into the bloodstream.
- Such liver delivery has been proposed to be particularly useful, for example, for expression of proteins that are missing in certain metabolic or hematological disorders, or that are effective in provoking immune responses (e.g., particularly antibody responses), for example against infectious agents or cancer cells.
- administered LNPs are delivered to and/or taken up by antigen-presenting cells (e.g., as may be present in skin, muscle, mucosal tissues, etc.); such administration may be particularly useful or effective for induction of T cell immunity (e.g., for treatment of infectious diseases and/or cancers).
- antigen-presenting cells e.g., as may be present in skin, muscle, mucosal tissues, etc.
- T cell immunity e.g., for treatment of infectious diseases and/or cancers.
- lipid nanoparticles can have an average size (e.g., mean diameter) of about 30 nm to about 150 nm, about 40 nm to about 150 nm, about 50 nm to about 150 nm, about 50 nm to about 130 nm, about 50 nm to about 110 nm, about 50 nm to about 100 nm, about 50 to about 90 nm, or about 60 nm to about 80 nm, or about 60 nm to about 70 nm.
- lipid nanoparticles that may be useful in accordance with the present disclosure can have an average size (e.g., mean diameter) of about 50 nm to about 100 nm.
- lipid nanoparticles may have an average size (e.g., mean diameter) of less than 80 nm, less than 75 nm, less than 70 nm, less than 65 nm, less than 60 nm, less than 55 nm, less than 50 nm, or less than 45 nm.
- average size e.g., mean diameter
- lipid nanoparticles that may be useful in accordance with the present disclosure can have an average size (e.g., mean diameter) of about 45 nm, 50 nm, 55 nm, 60 nm, 65 nm, 70 nm, 75 nm, 80 nm, 85 nm, 90 nm, 95 nm, 100 nm, 105 nm, 110 nm, 115 nm, 120 nm, 125 nm, 130 nm, 135 nm, 140 nm, 145 nm, or 150 nm.
- average size e.g., mean diameter
- lipids that form lipid nanoparticles described herein comprise: a polymer-conjugated lipid; a cationic lipid; and a helper neutral lipid.
- total polymer- conjugated lipid may be present in about 0.5-5 mol%, about 0.7-3.5 mol%, about 1-2.5 mol%, about 1.5-2 mol%, or about 1.5-1.8 mol% of the total lipids.
- total polymer-conjugated lipid may be present in about 1-2.5 mol% of the total lipids.
- the molar ratio of total cationic lipid to total polymer-conjugated lipid may be about 100:1 to about 20:1, or about 50:1 to about 20:1, or about 40:1 to about 20:1, or about 35:1 to about 25:1. In some embodiments, the molar ratio of total cationic lipid to total polymer-conjugated lipid may be about 35:1 to about 25:1.
- total cationic lipid is present in about 35-65 mol%, about 40-60 mol%, about 41-49 mol%, about 41-48 mol%, about 42-48 mol%, about 43-48 mol%, about 44-48 mol%, about 45-48 mol%, or about 46-49 mol% of the total lipids.
- total cationic lipid is present in about 47.0, 47.1, 47.2, 47.3, 47.4, 47.5, 47.6, 47.7, 47.8, 47.9 or 48.0 mol% of the total lipids.
- 11890709v1 24 Attorney Docket No.: 2013237-0929 [0164]
- total neutral lipid is present in about 35-65 mol%, about 40- 60 mol%, about 45-55 mol%, or about 47-52 mol% of the total lipids.
- total neutral lipid is present in 35-65 mol% of the total lipids.
- total non-steroid neutral lipid e.g., DPSC
- DPSC total non-steroid neutral lipid
- total non-steroid neutral lipid is present in about 5-15 mol%, about 7-13 mol%, or 9-11 mol% of the total lipids.
- total non-steroid neutral lipid is present in about 9.5, 10 or 10.5 mol% of the total lipids.
- the molar ratio of the total cationic lipid to the non-steroid neutral lipid ranges from about 4.1: 1.0 to about 4.9: 1.0, from about 4.5: 1.0 to about 4.8: 1.0, or from about 4.7: 1.0 to 4.8: 1.0.
- total steroid neutral lipid e.g., cholesterol
- total steroid neutral lipid is present in about 35- 50 mol%, about 39-49 mol%, about 39-46 mol%, about 39- 44 mol%, or about 39-42 mol% of the total lipids.
- total steroid neutral lipid e.g., cholesterol
- the molar ratio of total cationic lipid to total steroid neutral lipid is about 1.5:1 to 1: 1.2, or about 1.2: 1 to 1: 1.2.
- a lipid composition comprising a cationic lipid, a polymer-conjugated lipid, and a neutral lipid can have individual lipids present in certain molar percents of the total lipids, or in certain molar ratios (relative to each other) as described in WO 2018/081480, the entire contents of each of which are incorporated herein by reference for the purposes described herein.
- lipids that form the lipid nanoparticles comprise: a polymer- conjugated lipid (e.g., PEG-conjugated lipid); a cationic lipid; and a neutral lipid, wherein the polymer- conjugated lipid is present in about 1-2.5 mol% of the total lipids; the cationic lipid is present in 35-65 mol% of the total lipids; and the neutral lipid is present in 35-65 mol% of the total lipids.
- lipids that form the lipid nanoparticles comprise: a polymer-conjugated lipid (e.g., PEG-conjugated lipid); a cationic lipid; and a neutral lipid, wherein the polymer-conjugated lipid is present in about 1-2 mol% of the total lipids; the cationic lipid is present in 45-48.5 mol% of the total lipids; and the neutral lipid is present in 45-55 mol% of the total lipids.
- a polymer-conjugated lipid e.g., PEG-conjugated lipid
- lipids that form the lipid nanoparticles comprise: a polymer-conjugated lipid (e.g., PEG-conjugated lipid); a cationic lipid; and a neutral lipid comprising a non-steroid neutral lipid and a steroid neutral lipid, wherein the polymer-conjugated lipid is present in about 1-2 mol% of the total lipids; the cationic lipid is present in 45-48.5 mol% of the total lipids; the non-steroid neutral lipid is present in 9-11 mol% of the total lipids; and the steroid neutral lipid is present in about 36-44 mol% of the total lipids.
- a polymer-conjugated lipid e.g., PEG-conjugated lipid
- a cationic lipid e.g., PEG-conjugated lipid
- a neutral lipid comprising a non-steroid neutral lipid and a steroid neutral lipid
- a PEG-conjugated lipid is or comprises a structure as described in WO 2017/075531 (also described , or a some a PEG-conjugated lipid is or comprises 2-[(polyethylene glycol)-2000]-N,N-ditetradecylacetamide.
- a cationic lipid is or comprises a chemical structure selected from I-1 to I-10 of Table 1 herein or a derivative thereof.
- a cationic lipid is or comprises ((4-hydroxybutyl)azanediyl)bis(hexane-6,1-diyl)bis(2-hexyldecanoate).
- a neutral lipid comprises DSPC and cholesterol, wherein DSPC is a non-steroid neutral lipid and cholesterol is a steroid neutral lipid.
- lipid nanoparticles include one or more cationic lipids (e.g., ones described herein).
- cationic lipid nanoparticles may comprise at least one cationic lipid, at least one polymer-conjugated lipid, and at least one helper lipid (e.g., at least one neutral lipid).
- Figure 1 depicts a schematic of an exemplary drug manufacturing process 10.
- the process 10 may include the production of drug substance 12 followed by the production of drug product 14.
- Production of drug substance 12 may include in vitro transcription 16 followed by tangential flow filtration 18, followed by 0.2 micron filtration 20, followed by storage 22 at one or more temperatures in a range from about -20 deg C to about 8 deg C.
- Storage 22 may occur in a different location than the production of drug substance 12.
- drug subtance 12 may need to be transported to another location for production of drug product 14, which may include lipid nanoparticle (LNP) formation 24, followed by a second tangential flow filtration 26 process, followed by formulaion 28 and 0.2 micron filtation, folllowed by a second storage 30 process at one or more temperatures in a range from about -20 deg C to about 8 deg C.
- LNP lipid nanoparticle
- Figures 11-13 illustrate an exemplary LNP manufacturing process (for example, an RNA-LNP manufacturing process).
- Figure 11 illustrates an overview of exemplary manufacturing process 520 for a pharmaceutical-grade composition comprising RNA, according to aspects of the present disclosure.
- the process 520 may include the DNA transcription module 128, the first purification module 130, and the first bioburden reduction (or filtration) module 132, as previously described herein.
- the DNA transcription module 128, the first purification module 130, and the first bioburden reduction (or filtration) module 132 as previously described herein.
- the process 520 includes an exemplary manufacturing process for pharmaceutical-grade RNA comprising an in vitro RNA transcription followed by removal of components utilized or formed in the course of production by a purification process, and filtration to reduce bioburden (e.g., as illustrated in Figure 11).
- Optional in-process controls may also be completed depending on whether a hold step is performed.
- Figure 12 illustrates an overview of exemplary DNA template manufacture process 530 via a PCR-based process, according to aspects of the present disclosure.
- the process 530 includes an exemplary manufacturing process of a DNA template via a PCR-based process including the DNA transcription module 128, the first purification module 130, and the first bioburden reduction (or filtration) module 132, as described herein.
- a master mix preparation is made. Subsequently, forward primer and vector are added. The PCR-mix is transferred into a reagent reservoir and a PCR plate was filled. A PCR is completed comprising an initial denaturation, a denaturation step, an annealing step, a final extension step for 20-30 (e.g., 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) cycles and a hold step. The PCR products can be pooled and purified. Subsequently, the purified, pooled PCR product may be filtered and quality control tested.
- Fig. 12 illustrates which portions and/or steps of the process 530 are contained within each module (i.e., 128, 130, 132) of the process flow shown in Fig. 11.
- FIG. 13 illustrates an exemplary process 800 for manufacturing LNP compositions.
- steps 806, 808, and 810 (as well as equipment associated with those steps, as described herein) will 11890709v1 26 Attorney Docket No.: 2013237-0929 occur and/or be located in the LNP formation module 70, shown in Fig. 5.
- steps 812 and 814 (as well as equipment associated with those steps, as described herein) will occur and/or be located in the second purification module (i.e., the second tangential flow filtration module) 80, shown in Fig.8.
- the second purification module i.e., the second tangential flow filtration module
- step 816 (as well as equipment associated with step 816, as described herein) will occur and/or be located in the second bioburden reduction module (i.e., formulation) 90 shown in Fig. 9. Steps following 816 may occur in other modules and/or may occur at other facilities (or not at all). For example, as explained herein, freezing and warehousing may not be required in all embodiments.
- the produced compositions are prepared by combining lipids 810 with an aqueous preparation which carries an agent of interest (e.g., an active agent).
- the agent of interest is a nucleic acid (e.g., a nucleic acid therapeutic).
- the nucleic acid is an RNA (e.g., a therapeutic RNA); in many embodiments of this depicted process, a utilized RNA includes at least one open reading frame (ORF) which may, for example, encode a vaccine antigen, a replacement protein, an antibody agent, a cytokine, etc.).
- ORF open reading frame
- a vaccine antigen may be a cancer vaccine antigen or an infectious disease (e.g., viral) antigen.
- an RNA encodes a polypeptide that is or comprises a viral antigen such as a coronaviral antigen, such as a spike protein or portion thereof, or relevant variant of the foregoing (e.g., a SARS-CoV-2 spike protein or receptor binding domain thereof, for example, a prefusion stabilized variant thereof), e.g., as is utilized in one or more of mRNA- BNT162a1, mRNA-BNT162b1, mRNA-BNT162b2, mRNA-BNT-162c1, mRNA-1273, CVnCov, CVnCoV2, etc.).
- a viral antigen such as a coronaviral antigen, such as a spike protein or portion thereof, or relevant variant of the foregoing (e.g., a SARS-CoV-2 spike protein or receptor binding domain thereof, for example, a prefusion stabilized variant thereof), e.g., as is utilized in one or more of mRNA- BNT162a1, mRNA-B
- the RNA is prepared by in vitro transcription (e.g., of a DNA template which may, for example be a linear template such as a linearized plasmid or an amplicon).
- the process 800 may include LNP formation by adding lipids 810 to an RNA solution 806, as well as high impact mixing (for example, via impingement jet mixing), and stabilization.
- the RNA solution is an aqueous solution.
- the lipids 810 may include one or more of a cationically ionizable (sometimes referred to as “cationic” for simplicity) lipid, a phospholipid, a PEG-lipid, a sterol (e.g., a cholesterol) and an appropriate solvent (e.g., ethanol).
- LNP formation may be performed in presence of a buffer (e.g., a citrate buffer) 812.
- the buffer (e.g., a citrate buffer) 812 may be present in the RNA solution 806 prior to mixing with the lipids 810 (for example, via in-line dilution of the water-diluted RNA with the buffer (e.g., citrate buffer) 812 to form the aqueous solution of RNA 806). Stated otherwise, buffer (e.g., citrate buffer) 812 may be added to the RNA solution prior to mixing with the lipid solution 810. In some embodiments, the buffer (e.g., citrate buffer) 812 may also (or alternatively) be added to the mixture resulting from combining the lipid solution with the aqueous solution 806 (which, as depicted in Fig.
- step 808 includes reducing or eliminating the introduction of various impurities into the process and/or various solutions thereof, thereby 11890709v1 27 Attorney Docket No.: 2013237-0929 forming a first RNA-LNP preparation that includes LNP-encapsulated RNA.
- LNP formation 808 may include the adjusting of one or more process temperatures, process flow rates, and/or ratios of the buffers, solutions and/or suspensions.
- LNP formation may include independently flowing each of the aqueous solution and lipids 810 (for example, in a lipid solution) into a mixing unit.
- Each of the aqueous RNA solution 806 and lipid solution 810 may flow into the mixing unit under laminar flow conditions (to avoid the entrapment of gas bubbles (for example, ambient gas bubbles (i.e., primarily comprised of nitrogen)) within the flow).
- gas bubbles for example, ambient gas bubbles (i.e., primarily comprised of nitrogen)
- the process 800 may include buffer exchange and concentration of the first RNA-LNP preparation to form a second RNA-LNP preparation.
- the buffer exchange and concentration step 814 may be conducted with process parameters including, for example, a feed flow rate, for example within a range of 18 to 50 liter/min (LPM), a trans-membrane pressure (TMP), for example lower than 1200 mbar, a retentate pressure, for example within a range of 130 to 230 mbar, and a permeate pressure, for example within a range of 10 to 70 mbar.
- LPM trans-membrane pressure
- a retentate pressure for example within a range of 130 to 230 mbar
- permeate pressure for example within a range of 10 to 70 mbar.
- buffer exchange 814 of the first RNA-LNP preparation and concentrating the first RNA-LNP preparation are performed in alternating steps.
- a TRIS (i.e., tris(hydroxymethyl)aminomethane) buffer may be used.
- the buffer exchange 814 is conducted via diafiltration and the concentration is conducted via ultrafiltration.
- the diafiltration and/or the ultrafiltration are conducted via tangential flow filtration (TFF) (for example, in a tangential flow filtration unit and/or TFF skid).
- the tangential flow filtration is conducted using one or more jejunostomy tubes and/or one or more dip tubes configured to avoid introducing ambient gas bubbles into the second RNA-LNP preparation.
- a retentate may be recirculated to a feed tank using a dip tube comprising a first end submerged into filtration feed liquid in the feed tank to avoid introducing ambient gas bubbles into the filtration feed liquid.
- a filtration system for tangential flow filtration may be filled with a buffer to prevent introducing ambient gas bubbles into the second RNA-LNP preparation.
- the buffer exchange and concentration step 814 may include at least two buffer exchanges conducted via diafiltration alternating with at least two concentrations conducted via ultrafiltration.
- process temperatures may be maintained within a desired temperature range (for example, at or below about 25 deg C, or from about 2 deg C to about 25 deg C, or from about 15 deg C to about 25 deg C).
- pH may be continuously monitored (and may be maintained in a target range (for example, from about 7.0 to about 7.5, or from about 7.1 to about 7.3)) and shear may be maintained, for example in a range from about 2000 s ⁇ -1 to about 6000 s ⁇ -1, or from about 3000 s ⁇ -1 to about 5000 s ⁇ -1, or at about 4000 s ⁇ -1 (+/- 1%, 5%, and/or 10%).
- a recovery flush may be performed, during which time shear may be reduced to under about 2000 s ⁇ -1 (for example, under about 1500 s ⁇ -1, or under about 1000 s ⁇ -1).
- the pH may be maintained within a range from about 7.3 to about 7.5, for example following ultrafiltration and/or diafiltration.
- the pH of the first RNA-LNP preparation may be maintained at a pH that is higher than that of the cationic lipid (i.e., the cationic lipid in the lipid solution). Without wishing to be bound by any particular theory, it is proposed that doing so may reduce foaming of the liquid nanoparticles.
- the first and/or second RNA-LNP preparation(s) may be sterilized without introducing any ambient gas into the produced formulation.
- a relevant produced formulation may be a product for further manipulation, processing, packaging, and/or shipping.
- a produced formulation may be or comprise a drug product formulation, e.g., for administration to humans.
- one or more sterilization steps may be performed by sterile filtration; in some embodiments, sterile (or other) filtration may be conducted at a target pressure with substantially no pressure building up during the filtration process, for example at about 1.03 bar (or from about 1.02 bar to about 1.04 bar, from about 1.01 bar to about 1.05 bar, or from about 1.00 bar to about 1.1 bar).
- a utilized mixing unit may include one or more impingement jet mixing skids. Prior to mixing, the impingement jet mixing skids may be vented and/or flooded to remove gas bubbles from tubing of the impingement jet mixing skids.
- Mixing of the aqueous and lipid solutions may be performed within boundaries of the mixing unit and/or one or more impingement jet mixing skids.
- the aqueous solution does not contact the lipid solution.
- the flow rate ratio into the mixing unit of the aqueous solution to the lipid solution is about 3:1, or from about 2.75:1 to about 3.25:1, or from about 2.5:1 to about 3.5:1, and/or from about 2.5: to about 3:1.
- the mixing speed may be adapted to avoid entrapping ambient gas in the first RNA-LNP preparation.
- one or more mixing processes may include increasing the mixing speed gradually until a slight vortex has formed (for example, the mixing speed at or slightly above the point at which a visible vortex has formed), but below the mixing speed at which foam begins to form.
- the system for example, the impingement jet mixing skids, the TFF system (i.e., the tangential flow filtration unit), and/or components thereof
- the system may be assessed at one or more time points (e.g., monitored over time, e.g., periodically or continuously) for presence of gas bubbles.
- gas detection may be performed via one or more flowmeters (for example, via one or more Coriolis flowmeters), and/or by visual assessment (e.g., via the human eye and/or various types of cameras), viand/or other detection means.
- the aqueous solution and/or the lipid solution may be flowed into the mixing unit through one or more inlets disposed at a bottom portion of the mixing unit, and the resulting first RNA-LNP preparation may be released from the mixing unit through one or more outlets disposed at a top portion of the mixing unit.
- the mixing may be performed with a submerged mixer.
- foam may be generated during and/or after formation of the LNP-encapsulated RNA, and may be subsequently removed from the RNA-LNP preparation (for example, the foam may be removed from the first and/or second RNA-LNP preparation).
- the process 800 may include 0.2 ⁇ m filtration and/or the addition of sucrose and PBS for compounding.
- the process 800 may include bioburden reduction filtration (BBR) 816 following the buffer 11890709v1 29 Attorney Docket No.: 2013237-0929 exchange and concentration 814.
- Bioburden reduction filtration 816 may include filtering with 0.2 ⁇ m pore size (or for example, about a 0.22 ⁇ m pore size) or smaller filter.
- Bioburden reduction filtration 816 may also include using other pore sizes (for example, 0.45 ⁇ m pore size) as described herein.
- Bioburden reduction filtration 816 may also include filtering the post TFF-LNP suspension through a particulate reduction filter prior to filtering the suspension through (for example) the 0.2 ⁇ m pore size and/or 0.22 ⁇ m pore size bioburden reduction filter. In some embodiments, bioburden reduction filtration 816 may also include performing a filter recovery flush.
- the process 800 may include filling transport bags (for example, Flexsafe ® bags) with the filtered second RNA-LNP preparation, and performing a visual inspection 818 of the transport bags for gas bubbles.
- transport bags may be, for example 12 L bags, 50 L bags, 100 L bags, and/or other suitable bag sizes (e.g., depending on the batch size of the relevant RNA-LNP preparation), including bags that include a volume between 12 L and 50 L, and/or bags that include a volume between 50 L and 100 L.
- filling transport bags may include filling the bags to a volume in a range from about 30% to about 95%, or from about 40% to about 90%, or from about 50% to about 85%, or from about 60% to about 85% or from about 70% to about 85%, and/or other subranges therebetween of the total bag volume.
- Filled bags may be stored and/or shipped at a temperature in a range from about 1 deg C to about 15 deg C (for example, at about 2 deg C to about 10 deg C, or from about 2 deg C to about 8 deg C), or alternatively may be frozen to a temperature of about -70 deg C (for example, in a range from about -60 deg C to about -80 deg C).
- the bags Prior to shipment, the bags may be secured in or on racks and/or within or on any other suitable shelving or storage system so as to minimize movement, rupturing, and/or disruption of the bags during the transport to a fill and finish site.
- transport bags may be stacked in a specific manner using a stacking system on pallets that include shock absorbers.
- nitrogen with a positive pressure for example, from about 1-2 bars
- nitrogen with a positive pressure may be maintained in and around the environment in which the bags are kept and/or transported, in order to prevent ambient gas from entering the bags.
- the bags may be assessed for ambient gas content (e.g., visually inspected) 822 a second time.
- ambient gas bubbles that are discovered during such second assessment 822 may be removed (e.g., may be manually removed), or alternatively, the bag or bags that include ambient gas bubbles may be selectively discarded (for example, if the volume of ambient gas within a given bag has exceeded a threshold).
- sterile filtration 824 may be performed (i.e., the second RNA-LNP preparation). In some embodiments, such sterile filtration 824 may be performed after the preparation has been removed from the transport bags, but prior to being disposed within a collection vessel, reservoir, and/or fill tank.
- the material i.e., the filtered preparation
- the material may then be dispersed from the collection vessel, reservoir, and/or fill tank during aseptic fill and finish 826 (for example, to aseptically fill glass vessels with the drug product).
- 11890709v1 30 Attorney Docket No.: 2013237-0929 [0193]
- Visual inspection 828 may be performed on the filled glass vessels.
- the inspected and filled glass vessels, at step 830 of the process 800, may then be frozen, stored, warehoused and/or distributed, for example, to health care administration sites.
- filled glass vessels may be subjected to lyophilization or other drying process, so that drug product is transported and/or stored in a dry state (e.g., for subsequent resuspension).
- the fill and finish facility may be located in the same location as the LNP production facility, in which case fill and finish may be performed directly using Point of Fill filtration equipment (in which case the transport 820, bag filling and sealing, and one or more of the visual inspection steps 818, 822, 828 may not be required.
- the process 800 may include multiple transport steps 820, as well as additional visual inspection steps 818, 822, 828 if the various steps of the process 800 are performed at additional and/or other facilities (or alternatively, if transport is required within a single facility).
- Drug processing and preparation of lipid ethanol solutions, lipid nanoparticle (LNP) preparations (including RNA-LNPs), and other bioprocessing or chemical processing methodologies often require heating and cooling.
- the jacket of the present embodiments may be used as part of in vitro transcription (i.e., in connection with the bioreactors), tangential fill filtration, formulation, impingement jet mixing of lipids and RNA solutions, as well as during other processes.
- the jackets may be cooled, heated, and/or pressurized.
- the internal fluid flow may also be adjusted.
- the bottles and jacket rest on a table that has a magnetic stirrer (i.e., a “stirring plate” or “stirrer plate”).
- a bottom indentation in the bottle allows for impeller (or stirrer) to be seated on a hub around which it can rotate as a result of a magnetic coupling with a stirring plate in the table.
- the single use and/or multi-use bottles may be made from polypropylene, as well as other suitable materials.
- the jacket may be formed from stainless steel and/or other metallic or other materials that are sufficiently conductive to heat.
- the jacket may include a temperature control unit to maintain the internal temperature at the desired levels.
- the jacket may include one or more clamps located at the fluid inlets and outlets, which may be turned to enable thermal fluid flow into and out of the vessel.
- the jacket may include a fluid inlet and a fluid outlet, for routing cooling and/or heating fluids in and out.
- cooling and/or heating fluids that are used in connection with the jacket may include water, silicon oil, ethanol, and/or other thermal fluids.
- the jacket may include one or more sensors (for example, temperature probes such as RTDs, thermocouples, other types of temperature probes, pressure sensors, differential pressure sensors, flow sensors, etc.).
- the jacket includes two sides, shells, and/or halves such that the jacket may be opened and closed.
- one side, shell, or half of the jacket may be fixed to a table.
- the two shells may be connected via hinges such that one side, half, or shell rotates about the one or more hinges relative to the second side, half, or shell.
- the two sides, halves, and/or shells may be hinged into a closed position and attached together via one or more attachment means such as a latch, buckles, clasp, bayonet feature, and/or other suitable attachment means.
- the attachment means is non-magnetic so as to prevent any interference with the heating / cooling fluid and/or the contents within the bioprocessing vessel / bioreactor 11890709v1 31 Attorney Docket No.: 2013237-0929 and/or single-use bottles.
- one or more programmable limit switch (PLS) controllers may be used to control the magnetic stirred speed and check the temperature.
- the bottle may be composed of polypropylene and may be configured to accommodate internal pressures up to about 1 bar and/or in some embodiments, up to 5-10 bars.
- the bottle may have an internal volume of about 1 L to about 50 L, or from about 2 L to about 5 L or from about 10 L to about 20 L, or from about 20 L to about 50 L, or from about 2 L to about 20 L, or from about 4 L to about 50 L, or from about 5 L to about 20 L, or from about 5 L to about 10 L, and/or other subranges therebetween.
- the jacket is used to heat to temperatures in the vicinity of 35 deg C, or from about 30-40 deg C, while also being configured to cool to temperatures in a range from about 2 deg C to about 8 deg C.
- the jacket may be used to provide a wide temperature range, for example, temperatures in a range from 0 deg to 100 deg, or from about 1 deg C to about 99 deg C.
- the jacket is shaped (i.e., includes internal contouring and/or features) to match and/or accommodate the size, shape, and/or features of a particular single use bottle, bioreactor, and/or other bioprocessing vessel.
- the jacket may be used in connection with bioreactors and/or single use bottles that include one or more inlets at the top (for example, for adding lipids or other materials, as well as one or more additional openings for siphoning off portions of the mixture contained therein.
- the jacket may be used in connection with a bottle or vessel (for example, a bioreactor) that includes both a bottom stirrer and a top stirrer (for example, magnetically actuated stirrers disposed in the interior of the bottle or bioreactor with one or more corresponding electromagnetic inducers disposed within the bottom surface and/or top surface of the bottle and/or bioreactor, such that the electromagnetic inducers are magnetically coupled to the respective magnetic stirrer).
- the jacket may be used in connection with one or more drug production processes, for example in the preparation of a lipid-ethanol solution.
- the jacket may be used in connection with a tangential flow filtration process and/or in connection with a drug formulation process.
- the jacket can be used in any application where heating, cooling, and/or stirring are required.
- the jacket may be used in connection with a buffer preparation process.
- the jacket does not need to be cleaned after each use because the jacket includes a closed loop system for circulating one or more cooling or heating fluids, as described herein.
- the closed loop system(s) within the jacket are fluidly disconnected from the interior of the single-use bottle, bioreactor and/or bio-processing vessel. Accordingly, there is low to no risk of cooling/heating fluid from the closed-loop systems within the jacket from impurifying and/or otherwise contaminating the processes within the interior of the single use bottle / bioreactor.
- Each of the jackets of the present embodiments may be used to accommodate a number of processes with varying process parameters.
- the jacket in a first use, the jacket could be used to heat a bioreactor being used in an in vitro transcription process.
- the same jacket could be used to cool a single use bottle or vessel being used to mix ethanol and lipids.
- the jacket provides the desired conditions (i.e., temperatures) to the vessel positioned within in it via the temperature control unit (which also dictates the fluid pressure and flow rate).
- the inlet and/or outlet may include one or more sealing features (for example, rubber gaskets, O-rings, quick-connects, and/or other features) at the interface between the internal channels of the jacket and the fluid lines connected thereto.
- the jacket may include a pair of semi-circulate plates or outer casings disposed around the outer periphery of the jacket (for example, one semi-circulate plate or casing disposed around each 11890709v1 32 Attorney Docket No.: 2013237-0929 of the two jacket shells) to seal the channels (i.e., forming the radially outer walls of the channels).
- the semi- circulate plates or outer casings may be composed of insulating materials (i.e., rather than heat-conducting materials) so as to discourage heat transfer between the jacket / channels and the external environment.
- the outer casing may be composed of a metallic material such as stainless steel. In some embodiments, where heat transfer to the environment is more of a consideration (i.e., in connection with larger jackets) the outer casing may be composed of an insulting material such as a structural polymer or foam. According to aspects of the present embodiments, an important consideration includes selecting structural polymers or other insulating materials that are not reactive with the thermal fluids. As such, in some embodiments, the outer casing includes a composite structure that includes a metallic (i.e., stainless steel) inner layer and an insulating (i.e., polymer or foam) outer layer.
- the jacket may include one or more grooves to allow for tubing (to allow inlet / outlet flows) to be disposed therethrough, the tubing connecting an interior of the vessel, bottle, or bioreactor to an exterior of the vessel, bottle, or bioreactor.
- internal features of the jacket may be made via casting, forging, 3D printing / additive manufacturing, machining, fabrication, milling, CNC drilling, electrical discharge machining (EDM), and/or other suitable processes.
- the jacket may include an outer diameter of about 28 cm to accommodate a 10 L bottle / bioreactor.
- the jacket may include other dimensions and/or may accommodate a different size vessel or container.
- the jacket may be produced by starting with a stainless-steel cube and using one or more milling and / or computer numerical control (CNC) processes (for example, drilling using a 5-axis machine) to form the exterior and interior shapes including channels, protrusions, grooves, bores, etc.
- CNC computer numerical control
- the outer diameter may be smaller or larger to accommodate smaller or larger bottle sizes.
- the internal size and number of channels and/or channel passes may be scaled up and down in order to optimize the heating and/or cooling effectiveness.
- the jacket of the present invention may be used in connection with the drug manufacturing process (and various sub-processes and equipment used thereine) of Fig.2.
- the process may include in vitro transcription 50, a first tangential flow filtration process 60, LNP formation 70, a second tangential flow filtration 80, formulation 90, and fill and finish 100.
- the automated drug manufacturing process 40 of the present embodiments illustrated in Fig.
- a fully automated process occurring all at a single site (for example, at a single location within a single, contiguous footprint (i.e., production facility) comprising a total area of no more than about 8,000-12,000 square meters (for example, within an area comprising from about 100 square meters to about 12,000 square meters, or from about 200 square meters to about 10,000 square meters, or from about 300 square meters to about 8,000 square meters, or from about 200 square meters to about 6,000 square meters, or from about 500 square meters to about 3,000 square meters, or from about 1,000 square meters to about 5,000 square meters, or from about 1,500 square meters to about 4,000 square meters, or from about 750 square meters to about 3,500 square meters, or from about 500 square meters to about 1,750 square meters, and/or other subranges therebetween).
- a single site for example, at a single location within a single, contiguous footprint (i.e., production facility) comprising a total area of no more than about 8,000-12,000 square meters (for example, within an area comprising from about 100 square
- FIG. 1 illustrates several steps of the exemplary process 10 illustrated in Fig. 1 (such as transport between locations, and multiple storage (and thawing) steps)
- steps of the exemplary process 10 illustrated in Fig. 1 may be able to be eliminated.
- the process 40 is fully automated, less manpower is required to run the process.
- the process may be monitored 11890709v1 33 Attorney Docket No.: 2013237-0929 remotely, with no human intervention required to run the process once it has been supplied with electricity, a water supply, and/or the necessary input materials.
- Figures 3A and 3B illustrate perspective views of single use bottles 52 in a heating and cooling jacket 42, according to aspects of the present disclosure.
- Each of the heating and cooling jackets 42 may include first and second halves, portions or shells (i.e., “ shells”) 46, 48.
- Each of the first and second shell 46, 48 may be substantially semi-cylindrical such that the two shells 46, 48 together may form a jacket 42 (for example, a cylindrical jacket) around a single use bottle 52.
- each jacket 42 may be used in connection with single use bottles 52, as well as other bioprocessing vessels such as bioreactors, mixing vessels, reservoirs, etc.
- the first and second shells 46, 48 may come together along an interface 78 such that the jacket 42 tightly encapsulates the single use bottle 52.
- the jacket 42 may include a joining means 76 (for example, a clasp, latch, buckle, etc.) for securing the first and second shells 46, 48 together.
- a joining means 76 for example, a clasp, latch, buckle, etc.
- the first and second shells 46, 48 may hinge relative to one another about a hinge axis 68.
- the jacket 42 may include one or more hinges that are disposed co-linearly with the hinge axis 68, the one or more hinges not being visible in the illustration of Fig. 3B.
- each jacket 42 may be disposed on a support table 44 that may be used to support the jacket 42 and single use bottle 52.
- the support table 44 may also be used to actuate (for example, rotate) a stirrer or impeller 89 (shown in Fig. 5A) positioned in the bottom of the single use bottle 52 (or bioprocessing vessel 52).
- the table 44 may include local controls 64 and/or a control unit for controlling the speed of the stirrer.
- the table 44 includes a magnetic field generator and/or an inducer that is/are magnetically coupled to the stirrer 89, thereby enabling articulation of the stirrer (or impeller) 89 by the table controls 64.
- the single use bottle 52 may include a second stirrer or impeller 89 disposed near a top surface 62 of the single use bottle, the second stirrer also being disposed within the interior of the single use bottle 52.
- the second stirrer near the top of the single use bottle 52
- the first stirrer at the bottom of the single use bottle 52
- the second stirrer is instead coupled to a second magnetic field generator or inducer (not shown) located adjacent to (or in the vicinity of) the top surface 62 of the single use bottle 52.
- one half of the jacket 42 may be firmly attached to the table while the other half of the jacket 42 may be free to rotate about the hinge axis 68, thereby allowing the jacket 42 to be opened and closed for insertion and removal of the single use bottle 52.
- Firmly attaching one half of the jacket 42 to the table 44 helps to prevent the likelihood of the bottle 52 and/or jacket 42 from falling off of the table 44.
- each jacket 42 may include one or more inlets 66 and one or more outlets 54 for routing thermal fluids (such as silicone oil, water, mixes thereof, etc.) into and out of the jacket 42 for cooling and/or heating the contents of the bioprocessing vessel 42 (or single use bottle 42).
- the one or more inlets 66 are disposed at the bottom of the jacket 42 while the one or more outlets 54 are disposed at the top of the jacket 42, as shown in Fig. 3A. In some embodiments, the one or more inlets 66 are disposed at the top of the jacket 42 while the one or more outlets 54 are disposed at the bottom of the jacket 42, as shown in Fig. 3B.
- placing the one or more inlets 66 at the 11890709v1 34 Attorney Docket No.: 2013237-0929 bottom and the one or more outlets 54 at the top of the jacket 42 helps to encourage any trapped gas and/or air bubbles make their way out of the jacket 42, thereby preventing the gas(es) from building up in the interior of the jacket 42.
- Each of the one or more inlets 66 and one or more outlets 54 may be fluidly coupled (via any suitble means) to external tubing (not shown) for routing flows into and out of the jacket 42.
- each of the inlets 66 and outlets 54 may be fluidly connected to one or more channels and/or passageways or passages disposed within the interior of the jacket 42.
- Each jacket 42 may include one or more through-bores (for example, a small circular through-bore 72, a larger, elongated through-bore 82 (i.e., with rounded corners in some embodiments), as well as other through-bores, in various shapes and sizes) such that nipples 74, adaptors, couplings and/or other connections disposed within the single use bottle 52 may be connected to tubing that is positioned external to the jacket 42.
- Each single use bottle 52 may include one or more openings disposed in the top of each bottle (for example, a smaller opening 56 and a larger opening 58 for adding materials to and/or siphoning or suctioning materials from the the single use bottle 52).
- FIG. 4 illustrates a support table 44, according to aspects of the present disclosure.
- the table 44 may include a platform 84 for supporting the single use bottle 52 (and/or other bioprocessing vessel 52) as well as the local controllers 64 (i.e., for adjusting the speed at which the stirrer or impeller 89 (shown in Fig.
- FIG. 5A spins within the single use bottle 52).
- the platform 84 may also include a magnetic field generator or inducer (for example, located beneath the platform 84) for magnetically coupling to and actuating (i.e., rotating) the stirrer or impeller 89 within the single use bottle 52 (or bioprocessing vessel 52).
- Figures 5A and 5B illustrates details of a single use bottle 52 in a heating and cooling jacket 42, according to aspects of the present disclosure.
- Fig.5A illustrates a cross-section of a single use bottle 52 disposed within a jacket 42
- Fig.5B illustrates an interior perspective view of a jacket shell 48 (for example, a second jacket shell 48 with a small circular through-bore 72 and a larger elongated through-bore 82 disposed therethrough) according to aspects of the present embodiments.
- the jacket 42 may include a plurality of channels 86 that extend circumferentially around the jacket and interconnect with each other.
- each of the two shells 46, 48 that form the jacket 42 include an interconnected network of channels 86 or passages that extend circumferentially around (and within) the shell 46, 48 and connect to a channel that is longitudinally adjacent to each (for example, each channel 86 connects to the channel above and/or below it in the view illustrated in Fig.5A).
- the connections 124 between channels 86 are shown in Fig. 6.
- the plurality of channels 86 fluidly connect the fluid inlet 66 to the fluid outlet 54 within each of the shells 46, 48.
- each of the first shell 46 and the second shell 48 includes a flow circuit from the fluid inlet 66, to the plurality of channels 86, to the fluid outlet 54.
- the table 44 includes the magnetic field generator 88 (or inducer 88) disposed beneath the surface of the table, and in some embodiments may also include a control unit 92 (i.e., table control unit 92) for controlling the magnetic field generator 88 (or inducer 88), as illustrated in Fig. 5A.
- the control unit 92 may wirelessly (or via a wired connection) interface with other systems (for example, with a temperature control unit (TCU)).
- the table 44 may include a 11890709v1 35 Attorney Docket No.: 2013237-0929 plurality of supports 94 that extend vertically downward as well as laterally beneath the magnetic field generator 88 (or inducer 88) and control unit 92, so as to support those components.
- each of the first and second shells 46, 48 may include and/or be connected to one or more hinges 118 (for example, first hinge 118A and second hinge 118B, both being aligned with and centered on the hinge axis 68).
- the hinges 118A, 118B are illustrated as being attached to second shell 48 in Fig. 5B for illustration purposes, but would also connect to first shell 46.
- each hinge 118 of the plurality of hinges 118 may be located above the elongated through-bore 82.
- the plurality of channels 86 may include channels 86 that include different radial widths 122 and/or that are disposed at different radii from a bottle centerline 120.
- the jacket 42 may include varying widths 122 and varying radii from the centerline 120 at which the channels 86 are disposed in order to accommodate varying outer diameter profiles 110, 112, 114, 116 of the single use bottle 52.
- the single use bottle 52 may include one or more protruding portions 112, 116 that protrude radially outwardly beyond a nominal outer diameter 98 of the single use bottle 52 (i.e., which is also approximately a nominal inner diameter of the jacket 42).
- the single use bottle 52 may also include one or more recessed or concave portions 110 (when viewed externally) that extend radially inwardly to a radius that is less than the nominal inner diameter 98 of the jacket 42.
- the single use bottle 52 may also include one or more portions 114 (i.e., portions of the exterior surface of the bottle 52) that are disposed at a nominal outer diameter 98 of the single use bottle 52.
- the bottle 52 may include protruding portions 112, 116 to add structural robustness.
- the bottle 52 may include concave or recessed portions 110 to facilitate the easy of carrying the bottle (for example, by a forklift, human, or robot).
- the jacket may include corresponding protrusions and/or recesses.
- recesses 104 and 108 in the inner surface of the jacket 42 help to accommodate protrusions 112 and 116 of the bottle 52.
- protrusion 102 (which extends radially inward of the nominal inner diameter 98 of the jacket) helps to accommodate recessed portion 110 of the bottle 52.
- Portion 106 on the interior surface of the jacket 52 corresponds to portion 114 of the bottle 52, which is positioned at the nominal diameter 98.
- an outer diameter 96 of the jacket 42 may be substantially constant / uniform (for example, within standard manufacturing tolerances) while the inner diameter may intentionally vary (as described herein) around a nominal inner diameter 98 of the jacket 42.
- the plurality of channels 86 may include a constant (or uniform) outer radius about the centerline 120, and varying inner radii about the centerline 120.
- each of the channels runs circumferentially around the jacket (for example, around first jacket shell 46 and/or second jacket shell 48) and connect with adjacent channels 86 at connection channels 124 disposed at the ends of 11890709v1 36 Attorney Docket No.: 2013237-0929 each channel 86.
- a first plurality of channels 86A may include inner diameters that are more radially inward than the inner diameters of a second plurality of channels 86B, to accommodate bottles with varying outer diameters, as discussed herein in connection with Figs. 5A and 5B.
- the first and second shells 46, 48 may include a plurality of channel walls 142 defining the boundaries of each channel 86.
- Each channel 86 may also include a radially inner wall 140 that includes a semi-circulate contouring. As such, each channel extends in a semi-circular path around the jacket 42 and also includes a semi-circulare cross-section.
- an outer casing (not shown) that interfaces with lip 126 forms the radially outer walls of each channel 86.
- the outer casing may be welded, brazed, adhered, glued, compression fitted, mechanically attached, screwed into, and/or otherwise attached to the lip 126 and channel walls 142.
- the lip 126 extends around the exterior (i.e., radially outer) surface of each of the first and second shells 46, 48, and forms an interfacing surface for the outer casing to atttach to.
- the outer casing may be composed of metallic materials as well as one or more structural polymers that also acts as a thermal insulator such as polyethelene, polyvinylchloride, polypropylene, polyamide, as well as other suitable materials.
- the first and/or second shell 46, 48 may include a plurality of channels 138 with a shorter channel length 134 than the remainder of the channels 86, in order to accommodate and/or make space for the circular through-bore 72 and the elongated through-bore 82.
- the first and/or second shell 46, 48 may also include a vertically aligned manifold portion 136 for interfacing with the fluid inlet 66 (shown in Figs.3 and 5).
- Figures 7A and 7B illustrate a side view of a single use bottle 52 in a heating and cooling jacket 42 (Fig.7A) and a perspective view of a jacket 42 (for example, a second shell 48, Fig. 7B), according to aspects of the present disclosure.
- the heating and cooling jacket 42 in some embodiments, includes an outer diameter 51 that is larger than that of the single use bottle 52, and a height 47 that is smaller than that of the single use bottle 52.
- Fig. 7A the heating and cooling jacket 42, in some embodiments, includes an outer diameter 51 that is larger than that of the single use bottle 52, and a height 47 that is smaller than that of the single use bottle 52.
- the heating and cooling jacket 42 may include a sensor access port 49, for allowing a temperature sensor or other type of probe to be operatively coupled with the interior of the single use bottle 52 (i.e., thereby enabling the sensor to sense at least one operating parameter from the interior of the single use) while also allowing a wire or communication line 186 to extent outside of the heating and cooling jacket 42.
- the temperature sensor or other type of probe may be inserted into a protrusion 59 (shown in Fig. 7A).
- Figure 7C illustrates details of a heating and cooling jacket sensor access port 49, according to aspects of the present disclosure.
- the sensor access port 49 may include a first rounded portion 53, a second rounded portion 55, and an elongated portion 57.
- the elongated portion 57 separates the first rounded portion 53 from the second rounded portion 55.
- the first rounded portion 53 includes a larger radius than the second rounded portion 55.
- the first rounded portion 53 extends about 270 degrees (for example, from about 240 degrees to abuot 300 degrees, or from about 260 degrees to about 280 degrees) while the second rounded portion extends about 180 degrees (for example, from about 170 degrees to about 190 degrees).
- the first rounded portion 53 is centered about a location of a centerline of the sensor or probe when it is inserted into the protrusion 59 (which protrudes externally from the single use bottle 42).
- the elongated portion 57 and second rounded portion 55 enable the second shell 48 to be rotated about the single use bottle 52 when the jacket 42 is being closed such that the protrusion 59 is disposed through the sensor access port 49.
- elongated portion 57 and second rounded portion 55 make space for the protrusion 59, thereby enabling closure of the jacket 42 via rotation about the single use bottle 42.
- the sensor access 11890709v1 37 Attorney Docket No.: 2013237-0929 port 49 included only the first rounded portion 53 (i.e., and not elongated portion 57 or second rounded portion 55), closure of the heating and cooling jacket 42 may only be possible by moving the second shell 48 radially inward around the single use bottle 52, rather than rotating it into place, since the protrusion would interfere with an interior surface of the second shell 48.
- the sensor access port 49 may be dimensioned around the diameter and protrusion distance (i.e., the distance the protrusion 49 extends from the exterior of the bottle / vessel 52) of the protrusion 59, for each of the various jacket 42 sizes.
- the sensor access port 49 shown in Fig. 7C corresponds to a 10 L vessel / bottle 52, and may include, for example, a radius of about 9 mm for the first rounded portion 53, a radius of about 5.5 mm for the second rounded portion 55, and a third rounded portion (marked with “R3,0” in Fig. 7C) at a transition between the first rounded portion 52 and the elongated portion 57, the third rounded portion including a radius of about 3.0 mm.
- Figures 8A and 8B illustrate details of a fabricated / manufactured heating and cooling jacket (for example, of a second shell 48), according to aspects of the present disclosure.
- Figures 9A and 9B illustrate perspective views of single use bottles 52 in a heating and cooling jacket 42, according to aspects of the present disclosure.
- Figure 10 illustrates a system 150 for heating and cooling single use bottles 52 including the jacket 42, table 44, magnetic field generator 88 (or inducer 88), table control unit 92, fluid inlet 66, and fluid outlet 54, according to aspects of the present disclosure.
- the system 150 may include a pump 144 for pressurizing one or more cooling and/or heating fluids, as well as a pump suction line 170 and a pump discharge line 146.
- the system may include one or more temperature handling units 148 disposed fluidly downstream of the pump discharge line 146.
- the temperature handling unit 148 includes the capability to both heat and cool fluids (for example, thermal fluids such as silicon oil, water, glycol, water-glycol mixes, etc.).
- thermal fluids for example, thermal fluids such as silicon oil, water, glycol, water-glycol mixes, etc.
- the density of the thermal fluid does not exceed 1 kg/dm ⁇ 3.
- the thermofluid includes a water-glycol mixture or a water-ethylene-glycol mixture.
- a water-glycol mixture is used as the thermal fluid.
- the thermal fluid includes a pH in a range from 6.0 to 8.5.
- the temperature handling unit may include a heat exchanger that is in thermal communication with the fluid line(s) as well as both a heat source and a cooling source.
- the heat source may include a heater that uses liquid fuel as a heat source.
- the heat source may additionally or alternatively include an electric heater (for example, an induction heater or a resistance heater).
- the cooling source may include a refrigeration unit (i.e., a cooling or coolant circuit) and/or a Peltier device (which is electrically powered and can act as both a heating source and cooling source).
- the temperature handling unit 148 may include one or more expansion vessels or reservoirs for storing thermal fluids.
- the temperature handling unit 148 may include an evaporator upstream of a compressor, upstream of a condenser that is in turn positioned upstream of the evaporator.
- the temperature handling unit may also include one or more motors (i.e., one or more stepper motors), one or more pumps, stepper valves, and/or additional heat exchangers (for example, integrated into the evaporator and/or condenser).
- the system 150 may include one or more inlet lines (for example, first inlet line 152 and second inlet line 154) at the exit of the temperature handling unit 148 for delivering cooling / heating fluid to the inlets 66 of the jacket 42.
- a Huber Unistat 410, 410(w), or equivalent may be used in connection with heating and cooling of smaller jackets (i.e., 1 L, 2 L, 5 L, 10 L vessels) while a Unistat 530(w), 705(w) or equivalent may be used for large jackets (10 L, 20 L, 50 L vessels).
- the system may include one or more fluid outlet lines (for example, fluid outlet lines 158 and 160) for connecting the fluid outlets 54 to a downstream reservoir 166, which may be fluidly connected to the pump suction line 170.
- the system may include one or more valves 168 disposed in the pump suction line 170, as well as in other locations throughout the system.
- a single line (for example, in connection with a T positioned at or near the two fluid outlets 54) may be used to delivery fluid to the reservoir 166 instead of the first and second fluid outlet lines 158, 160.
- a single fluid inlet 66 and a single fluid outlet 54 are shown.
- each of the first shell 46 and the second shell 48 will have its own fluid inlet 66 and fluid outlet 54, as shown in Fig. 3A, and as disclosed herein.
- the system 150 may include one or more robots (for example, a first robot 162 and a second robot 164) for moving various components of the system as needed including but not limited to, the single use bottle 52, the jacket 42, as well as other components.
- the temperature control unit 172 may act as more of an overall control system as it is interacting with, and potentially sending commands to, various components that do not function in a way that affects the temperature of the interior of the single use bottle 52, etc.
- the temperature control unit (TCU) 172 may be controlled via a PLS / programmable limit switch, (for example, a Siemens PCS7 or equivalent with Ethernet / TCP/IP communication capabilities, for example, capable of operating with one or more of the following telecontrol protocols: SINAUT ST7, DNP3, Modbus RTU, IEC 60870-5-101, and IEC 60870-5-104).
- the pump 144 may be located fluidly downstream of the temperature handling unit 148.
- Figure 11 depicts an overview of an exemplary manufacturing process for a pharmaceutical- grade composition comprising RNA that may include the use of the jacket 42, according to aspects of the present disclosure, and as discussed above.
- Figure 12 illustrates an overview of exemplary DNA template manufacture process via a PCR-based process that may include the use of the jacket 42, according to aspects of the present disclosure, and as discussed above.
- 11890709v1 39 Attorney Docket No.: 2013237-0929
- Figure 13 illustrates an exemplary process for manufacturing LNP compositions that may include the use of the jacket 42, according to aspects of the present disclosure, and as discussed above.
- the method 900 may include setting initial process conditions via the temperature control unit such that thermal fluid temperature, pressure, and/or flow rate are adjusted to deliver the desired heating and/or cooling to the bioprocessing vessel 42.
- the method 900 may include determining the flow rate, pressure, and/or temperature at which the thermal fluid will be circulated within the jacket 42.
- the method 900 may include measuring at least one parameter within the bioprocessing vessel 42.
- the method 900 may include setting a secondary set of process conditions (for example, different conditions than those set in step 910).
- the method 900 may include determining the required flow rate, pressure and/or temperature at which the thermal fluid will be circulated within the jacket 42 to provide the secondary process conditions (i.e., temperature) within the bioprocessing vessel 52.
- the method 900 may include measuring at least one process parameter within the bioprocessing vessel 52.
- the method 900 may include repeating steps 910 through 920 (for example, including adjusting the conditions to third, fourth, fifth, etc. sets of conditions) until the overall bioprocessing process is complete.
- the first set of process conditions may include a temperature that is above room temperature while the second set of process conditions (916) may include a temperature that is below room temperature, and vice versa.
- a temperature above room temperature may include a temperate in a range from about 26 deg C to about 30 deg C, or from about 30 deg C to about 34 deg C, or from about 35 deg C to about 40 deg C, or from about 32 deg C to about 37 deg C, or from about 26 deg C to about 40 deg C.
- the temperature set at steps 910 and 912 may be at least 15 deg C different than the temperature set at steps 916 and 918. In some embodiments, the temperature set at steps 910 and 912 may be at least 20 deg C different than the temperature set at steps 916 and 918. In some embodiments, the temperature set at steps 910 and 912 may be at least 25 deg C different than the temperature set at steps 916 and 918.
- the temperature set at steps 910 and 912 may be at least 30 deg C different than the temperature set at steps 916 and 918.
- the system described herein may employ the same thermal fluid (water, silicon oil, mixes thereof, etc.), for both cooling and heating.
- the steps illustrated in Fig. 14 may be performed in a different order than what is shown.
- the method 900 may include additional steps (i.e., steps not shown in Fig. 14).
- not every step shown in method 900 is performed.
- one or more steps are performed simultaneously rather than sequentially.
- FIG. 15 illustrates a heating and cooling jacket 42 in an open position, according to aspects of the present disclosure.
- the jacket may include the first shell 46, the second shell 48, the protrusion 102, and may be positioned on the table 44.
- the table may include an opening 172 (for example, a circular opening positioned in the center of the top surface of the table 44) such that the magnetic field generator 88 may be disposed therethrough so as to allow for unhindered electromagnetic coupling with the stirrer/impeller 89.
- the jacket 42 may include a single hinge 118 (i.e., or multiple hinges 118A, 118B as shown in Fig. 5B).
- the first shell 46 is rigidly coupled to the table 44 such that it is concentrically disposed about the opening 172 (i.e., the first shell 46 is positioned so that it is concentric (i.e., radially outward of) half of the circular opening 172; in a closed position, both the first shell 46 and the second shell 48 are concentric about the circulate opening 172).
- the hinge 118 allows the second shell 48 to swing open and closed such that vessels may be placed within, and removed from, the jacket 42. As shown in Fig.
- the latch 76 (which may be coupled to the first shell 46) may be used in connection with the catch 170 (positioned on the second shell 48) to receive the latch and keep the jacket 42 secured around the vessel / single use bottle when in operation.
- Each of the first and second shells 46, 48 may include a plurality of holes 174 positioned within respective circumferential faces 178 (or surfaces 178).
- the plurality of holes act as thermal expansion features.
- dowels (not shown) may be positioned in the plurality of holes 174 to help keep the first and second shells 46, 48 positioned relative to each other (for example, via compression fitting of the dowels within the holes 174).
- the dowels may also help to evenly distribute axial or verticle loading acting on one or both of the first and second shells 46, 48.
- the table 44 includes a plurality of feet 176 (for example, four (4) feet that are vertcally adjustable via threading that screws into and out of a bottom surface of the table such that the table may be leveled).
- Figure 16 illustrates a heating and cooling jacket 42 in a closed position with the first shell 46 secured to the second shell 48 via latch 76, according to aspects of the present disclosure.
- the outer casing 180 (for example, a metallic (e.g., stainless steel) outer casing) is visible in Fig. 16. In the embodiment of Fig.
- the jacket 42 and/or system 150 may include a communication wire 186 for communicating signals from the one or more sensors (for example, temperature sensors) that are disposed in the interior of the jacket 42, to the temperature control unit 172 and/or the temperature handling unit 148.
- the heating and cooling jacket 42 illustrated in Fig. 17 is dimensioned and sized to be used with a 2 L fluid vessel, bioprocessing container, and/or single use bottle.
- Figure 18 illustrates a heating and cooling jacket 42 in a closed position without outer casings 180, according to aspects of the present disclosure. In the embodiment of Fig.
- the diameters, heights, and spacings are all in millimeters (mm).
- the jacket diameters (i.e., inner and outer diameters) and heights of the heating and cooling jackets 42 scale up and down approximately in concert with the bottle volume (i.e., the approximate calculated volume that can be derived from the jacket diameter and height scales up and down approximately linearly with the volume of the corresponding bottle that the jacket is configured for).
- the channel diameters and channel spacings by contrast, remain within narrow ranges (and do not scale up and down approximately linearly with the bottle volume). For example, as the bottle volume increases from 1 L to 20 L, the channel diameter of the corresponding heating and cooling jacket increases from 12 mm to 15 mm while the channel spacing increases from 7 mm to 10 mm.
- the number of passes is (i.e., number of vertically stacked channels, each channel extending circumferentially) is a function of the heating and cooling jacket height, the channel diameter and channet spacing.
- the geometry and dimensions of the 50 L heating and cooling jacket 42 are qualitatively different than jackets 42 sized for other volumes, due to manufacturability.
- Bottle Channel Channel Number of Inner Outer Volume Diameter Spacing Passes Height Diameter Diameter [0230] Referring still to Table 1, the disclosed dimensions are for exemplary purposes only. Heating and cooling jackets 42 according to the present embodiments may be designed and manufactured using other sizes and dimensions.
- heating and cooling jackets with channel diameters that are significantly larger than those of the present embodiments are likely to have reduced cooling and/or heating effectiveness since a larger portion of the thermal fluid flows through the radially outer portion of the channels (i.e., closer to the outer casing and away from the bioprocessing vessel 52, which is where the heating and cooling is needed) thereby resulting in a relative loss of 11890709v1 42 Attorney Docket No.: 2013237-0929 convective heating/cooling to the bioprocessing vessel 52.
- the smaller channel spacing (for example, 7mm) in the jacket 42 used with the 1 L bottle helps to maximize the number of channel passes 86 that can fit within the smaller height of the 1 L bottle.
- heating and cooling jackets 42 sized for 1 L bottles up to and including 20 L jackets may be produced by milling of metal while the 50 L jacket 42 may be turned from a tube.
- sheet metal may be rounded to form the cylindrical walls of the 50 L jacket 42 while the planar walls (for example, horizontally oriented and/or radially oriented walls that form and define the channels may be made of flat steel (i.e., sheets of steel) that are machined to share and welded on. Therefore, the cross-sections of the channels of the 50 L jacket 42 may be, for example, rectangular while the cross-sections of the 1 L-20 L jackets 42 may be at least partially circular (for example, semi-circular) as described herein.
- FIG. 25 illustrates an interior view of a 50 L heating and cooling jacket 42, according to aspects of the present disclosure.
- Figure 26 illustrates a cross-sectional view of a 50 L heating and cooling jacket 42, according to aspects of the present disclosure.
- Figure 27 illustrates a perspective view of a 50 L heating and cooling jacket 42, according to aspects of the present disclosure. In the 50 L embodiment(s) shown in Figs.
- the heating and cooling jacket 42 may include a total of 8 passes or channels including six (6) interior channels 194 and two end channels 196 disposed above and below the interior channels 194.
- Each of the end channels 196 in some embodiments, include a larger channel height 204 than the channel height 202 of the interior channels 194 (i.e., so as to accommodate and make space for the one or more inlets 66 and/or the one or more outlets 54, which are used for routing cooling and/or heating fluid into and out of the heating and cooling jacket 32).
- the heating and cooling jacket 42 configured to accommodate 50 L fluid vessels 52 may include cooling channels with a larger height than width. Cooling channels with a larger channel height 202, 204 than width 208, according to the present disclosure, help to improve cooling and/or heating effectiveness due to the contact surface area between the heating and cooling jacket 42 and the single use bottle 52 / bioreactor 52 being maximized as a ratio of the channel volume.
- the heating and cooling jacket 42 may include one or more fluid drains 198 (for example, to allow gravity draining of thermal fluids out of the heating and cooling jacket 42) as well as convex contouring 206 disposed in an interior surface of the heating and cooling jacket 42 to match the concave contouring of a single use bottle (or fluid vessel).
- Figure 28 illustrates a front view of a mobile bioreactor unit 200 with a heating and cooling jacket 42, according to aspects of the present disclosure.
- Figure 29 illustrates a side view of a mobile bioreactor unit 200 with a heating and cooling jacket 42, according to aspects of the present disclosure.
- Figure 30 illustrates a rear view of a mobile bioreactor unit 200 with a heating and cooling jacket 42, according to aspects of the present disclosure.
- the mobile bioreactor unit 200 of the present disclosure may include a cart 230 with legs (for example, four legs) and wheels 224 coupled to the legs via castors 222 to enhance maneuverability of the mobile bioreactor unit 200.
- the mobile bioreactor unit may include a vertically oriented frame 216 for supporting brackets 210 used to support valves (for example, pneumatic valves 234 (shown in Fig.31)) as well as one or more holders 212 for supporting and/or housing a filter capsule 248 (shown in Fig. 31).
- the cart 230 may be used to support both a 50 L single use bottle 52 / bioreactor 52 (and also smaller size fluid vessels 52) as well as the heating and cooling jacket 42. The cart 230 enables transportability of the mobile bioreactor unit 200.
- the mobile bioreactor unit 200 may also include a plurality of supports 94 that extend vertically downward as well as laterally beneath a platform on which the single use bottle / bioreactor 52 and jacket 42 are positioned, for example, to hold a magnetic field generator 88 (shown in Fig. 10).
- mobile bioreactor unit 200 (or system) may include a connection point 220 (for example, a flange 220) for disconnecting the wheels 224 and castors 222 from the legs of the cart 230 such that the cart 230 may be positioned securely on a scale 258 (for example, as shown in Fig. 34).
- the cart 230 may include load cells 256 in place of the wheels and castors 224, 222 (as shown in Fig. 33), such that the mobile bioreactor system 200 (including the contents of the single use bottle / bioreactor 52) may be weighed.
- the mobile bioreactor system 200 may also include a support plate 218.
- the support plate 218 may be supported by the frame 216, and itself may be used to support a control cabinet that may contain the control units for: the magnetic stirrer 89 / magnetic field generator 88, one or more pressure sensors, temperature probes and/or valves 234 (shown in Fig. 31).
- the single use bottle 52 / bioreactor 52 may also include one or more handles 214 disposed at or near a top end or surface of the single use bottle / bioreactor 52.
- the mobile bioreactor system 200 may include a grounding lug 226 to enable electrical grounding of the mobile bioreactor system 200 via a wire connected thereto, to enable use of the mobile bioreactor system 200 with ethanol and other potentially hazardous materials, while maintaining safe use. 11890709v1 44 Attorney Docket No.: 2013237-0929 [0236]
- Figure 31 illustrates a front view of a mobile bioreactor unit 200 with a heating and cooling jacket, according to aspects of the present disclosure.
- the mobile bioreactor unit 200 may include an ethanol supply 242 fluidly coupled upstream of a pump 244 (for example, an ATEX pump) for routing ethanol into the single use bottle 52 / bioreactor 52 via an ethanol supply line 264.
- a first vent line 260 may be coupled to the ethanol supply line 264 (and may further be connected to a filter (i.e., an EKV filter)) to allow air to be removed from the ethanol supply line 264 upstream of where ethanol enters the single use bottle 52 / bioreactor 52.
- a filter i.e., an EKV filter
- the mobile bioreactor unit 200 may also include an RNA solution and/or drug substance outlet line 246, for delivering an RNA solution or other biological solution to a downstream process (for example, to a tangential flow filtration module, a t-mixer, a y-mixer, an LNP formation module, a lipoplex formation module, a liposome formation module, another bioreactor, a 0.2 ⁇ m filtration module, and/or other types of downstream processes.
- the drug substance outlet line 246 may include a filter 248 (i.e., an EKV filter) for removing air from the biological solution via a second vent line 262.
- the mobile bioreactor unit 200 may also include an air inlet line / air supply line 228 and an air exhaust line / air outlet line 232, each with a valve 234 (i.e., a pneumatic valve) disposed therein, and each being supported by a respective support bracket 210.
- a valve 234 i.e., a pneumatic valve
- each of the air inlet line / air supply line 228 and the air exhaust line / air outlet line 232 may include one or more filters 238, 240 disposed therein.
- the mobile bioreactor unit 200 may also include: a thermal fluid supply line 254 for routing thermal fluid into the heating and cooling jacket 42 from the temperature control unit (TCU), a TCU return line 252 for routing thermal fluid from the heating and cooling jacket 42 back to the TCU, a magnetic field generator 88 supported by the cart 230, a magnetic stirrer 89 electromagnetically coupled to the magnetic field generator 88 and positioned within the single use bottle 52 / bioreactor 52, and a switch box 250 for electrically coupling the mobile bioreactor unit 200 to an external power supply.
- TCU temperature control unit
- TCU return line 252 for routing thermal fluid from the heating and cooling jacket 42 back to the TCU
- a magnetic field generator 88 supported by the cart 230
- a magnetic stirrer 89 electromagnetically coupled to the magnetic field generator 88 and positioned within the single use bottle 52 / bioreactor 52
- a switch box 250 for electrically coupling the mobile bioreactor unit 200 to an external power supply.
- Figure 32 illustrates a perspective view of a mobile bioreactor unit 200 without a heating and cooling jacket, and including the cart 230, grounding lug 226, brackets 210, holders 212, jacket 42, single use bottle / bioreactor 52, and other features, as described herein and according to aspects of the present disclosure.
- one of the first shell 46 and the second shell 48 (shown in Figs.25-27) forming the heating and cooling jacket 42 may be rigidly coupled to the planar top surface of the cart 230, for example, via welding, brazing, bolt, screw, nut, glue, epoxy, adhesion, mechanical coupling, and/or other suitable attachment means.
- the shell that is not rigidly coupled to the cart 230 may be rotated into place such that the heating and cooling jacket 42 surrounds the single use bottle 52 / bioreactor 52.
- the jacket 42 has been described primarily in connection with the use of single use bottles 52.
- heating and cooling jacket 42 of the present embodiments may be used in connection with any other number of bioprocessing and/or chemical processing vessels including (but not limited to) multi-use or single use bioreactors, tangential flow filtration (TFF) equipment such as TFF cassettes, mixing vessels, reservoirs, other types of filtration devices, thawing chambers, refrigeration units (for example, in place of refrigeration units), and/or in connection with other types of equipment.
- TFF tangential flow filtration
- the heating and cooling jacket 42 of the present embodiments may be used in connection with: one or more bioreactors during in vitro transcription 50; one or more TFF cassettes during a first and/or a second tangential flow filtration process 60, 80; a mixing vessel such 11890709v1 45 Attorney Docket No.: 2013237-0929 as a lipid – RNA impingement jet mixer and/or a lipid-ethanol mixing vessel during LNP formation 70; filtration for bioburden reduction during formulation 90, and/or during in situ storage and/or warehousing (i.e., to provide flexibility in allowing those steps to occur in place rather than having to have separate processes and/or locations for performing those functions).
- the heating and cooling jacket 42 of the present embodiments may be used in connection with with temperature control of an LNP batch, for the cooling of buffers, for cooling in the formulation 90 area, and for cooling in the fill and/finish area 100, as well as in connection with other potential applications.
- the jacket 42 and bioprocessing vessel 52 may be cylindrical.
- the jacket 42 and bioprocessing vessel 52 may be substantially square or cube-shaped with rounded corners.
- the bioprocessing vessel 52 may be substantially square or cube shaped with rounded corners while the outer shape of the jacket 42 may be substantially cylindrical, as shown in Figs.9A and 9B.
- the bottles / vessels 52 may include a substantially rectangular cross-section, and the jacket 42 may be correspondingly contoured and/or shaped to accommodate the dimensions of the vessel 52.
- each of the vessel 52 and jacket 42 may include other shapes.
- the heating and cooling jacket 42 may be used in connection with a mobile bioreactor system, as described herein. Equivalents [0239] Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Therefore, the scope of the present invention is not intended to be limited to the above Description. 11890709v1 46
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Engineering & Computer Science (AREA)
- Life Sciences & Earth Sciences (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Organic Chemistry (AREA)
- Zoology (AREA)
- Wood Science & Technology (AREA)
- General Engineering & Computer Science (AREA)
- Biomedical Technology (AREA)
- General Health & Medical Sciences (AREA)
- Genetics & Genomics (AREA)
- Biochemistry (AREA)
- Biotechnology (AREA)
- Microbiology (AREA)
- Sustainable Development (AREA)
- Clinical Laboratory Science (AREA)
- Thermal Sciences (AREA)
- Physics & Mathematics (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Analytical Chemistry (AREA)
- Mechanical Engineering (AREA)
- Apparatus Associated With Microorganisms And Enzymes (AREA)
Abstract
A system for heating and/or cooling a vessel includes: a heating and cooling jacket disposed around the vessel and including (1) a plurality of channels configured to allow a thermal fluid to flow therethrough, (2) at least one fluid inlet, and (3) at least one fluid outlet, each fluidly coupled to the plurality of channels; and a temperature handling unit fluidly coupled to the jacket and configured to selectively heat and cool the thermal fluid.
Description
Attorney Docket No.: 2013237-0929 HEATING AND COOLING JACKET FOR FLUID VESSELS Cross-Reference to Related Applications [0001] This application claims priority to U.S. Provisional Patent Application No. 63/458,019, filed April 7, 2023; and U.S. Provisional Patent Application No. 63/464,990, filed May 9, 2023, the title of each of which is “SINGLE USE DRUG PRODUCTION BOTTLES,” and the content of each of which is incorporated herein by reference in its entirety. Background [0002] Bioprocessing and chemical processing protocols often face challenges due to needing to meet critical process parameter levels (for example, temperatures) while not risking contamination of the process itself. In addition, the handling of bioprocessing and chemical processing vessels or containers increases the likelihood that disruption will be caused with the underlying process. Options for the cooling of chemical and biological processing equipment are currently limited. Summary [0003] The present disclosure provides technologies relating to a jacket that may be used in connection with the heating and cooling of bioreactors, and other biological and/or chemical processing vessels and containers. In some embodiments, the jacket may be used in connection with single use drug production bottles. In some embodiments, the jacket may be placed around a bioprocessing or chemical processing vessel, enabling the vessel to remain in situ. [0004] In one aspect, the present disclosed embodiments are directed to a system for heating and/or cooling a vessel includes: a heating and cooling jacket disposed around the vessel and including (1) a plurality of channels configured to allow a thermal fluid to flow therethrough, and (2) at least one fluid inlet and (3) at least one fluid outlet, each fluidly coupled to the plurality of channels; and a temperature handling unit fluidly coupled to the jacket and configured to selectively heat and cool the thermal fluid. [0005] In some embodiments, the fluid comprises at least one of water and silicon oil. [0006] In some embodiments, the vessel comprises at least one of a bioprocessing vessel and a chemical processing vessel. [0007] In some embodiments, the at least one fluid inlet comprises a first fluid inlet and a second fluid inlet, the at least one fluid outlet comprises a first fluid outlet and a second fluid outlet, the first fluid outlet being fluidly coupled to the first fluid inlet via a first fluid stream within the jacket, and the second fluid outlet being fluidly coupled to the second fluid inlet via a second fluid stream within the jacket, and the first fluid stream and the second fluid stream are fluidly disconnected within the jacket. [0008] In some embodiments, the system includes at least one pump disposed fluidly upstream of the temperature handing unit; and at least one reservoir disposed fluidly upstream of the at least one pump. [0009] In some embodiments, the system includes a temperature control unit (TCU) communicatively coupled to both the temperature handling unit and the at least one pump. 11890709v1 1
Attorney Docket No.: 2013237-0929 [0010] In some embodiments, the system includes at least one support table for supporting the vessel and the jacket, wherein the at least one support table comprises at least one of a magnetic field generator and an inducer configured to be magnetically coupled to, and to cause rotation of, a magnetic stirrer disposed within the vessel. [0011] In some embodiments, the vessel comprises a bioprocessing vessel comprising at last one of a bioreactor and a single use bottle. [0012] In some embodiments, the vessel comprises at least one concavity. [0013] In some embodiments, the jacket comprises at least one protrusion comprising contouring that corresponds to the at least one concavity of the vessel, the at least one protrusion protruding radially inward of a nominal inner diameter of the jacket. [0014] In another aspect, the present disclosure is directed to: a heating and cooling jacket comprising: a first semi-cylindrical shell comprising a first plurality of channels configured to allow a thermal fluid to flow therethrough, the first shell comprising a first fluid inlet and a first fluid outlet, each fluidly coupled to the first plurality of channels; and a second semi-cylindrical shell comprising a second plurality of channels configured to allow the thermal fluid to flow therethrough, the second shell comprising a second fluid inlet and a second fluid outlet, each fluidly coupled to the second plurality of channels; wherein the first and second shells form the jacket and are configured to both heat and cool, via the thermal fluid, at least one object disposed within the jacket. [0015] In some embodiments, the jacket includes at least one hinge coupling the first shell to the second shell, wherein in a closed position, the jacket comprises a substantially cylindrical shape. [0016] In some embodiments, each of the first and second pluralities of channels comprises a plurality of adjacent channels, each of which extends circumferentially around the respective first or second shell, and each of the channels comprises a substantially semicircular cross-section. [0017] In some embodiments, each of the first shell and the second shell comprises an outer casing that defines a radially outer wall of the channels, and the outer casings are composed of at least one structural polymer material. [0018] In some embodiments, each of the first and second shells are composed of stainless steel. [0019] In some embodiments, each of the first and second pluralities of channels comprise channels with a uniform outer diameter; and each of the first and second pluralities of channels comprise at least one channel that is positioned at a different inner diameter than at least one other channel within the same shell. [0020] In some embodiments, at least one of the first shell and the second shell comprises at least one through-bore that connects an interior of the shell to an exterior of the shell. [0021] In some embodiments, the jacket includes at least one sensor access port disposed in the first shell and/or the second shell. The sensor access port may include a first rounded portion, a second rounded portion, and an elongated portion disposed between the first rounded portion and the second rounded portion. 11890709v1 2
Attorney Docket No.: 2013237-0929 [0022] In another aspect, the present disclosure is directed to: a method of heating or cooling an object comprising: providing a cooling and heating jacket comprising a plurality of channels configured to allow flow of a thermal fluid therethrough; positioning at least one object to be heated or cooled within the jacket; circulating the thermal fluid through the plurality of channels such that the object is heated or cooled to a first temperature for a first period of time; and circulating the thermal fluid through the plurality of channels such that the object is heated or cooled to a second temperature for a second period of time; wherein at least one of the first temperature and the second temperature is below room temperature, and wherein at least one of first temperature and the second temperature is above room temperature. [0023] In some embodiments, there is at least a 15 deg C difference between the first temperature and the second temperature, at least one of the first temperature and the second temperature is in a range from about 1 deg C to about 17 deg C, and at least one of the first temperature and the second temperature is in a range from about 26 deg C to about 40 deg C. [0024] In some embodiments, the jacket is configured to heat and cool the object to temperatures in a range from about 1 deg C to about 99 deg C. [0025] In some embodiments, the vessel comprises at least one of: (1) one or more bioreactors during in vitro transcription, (2) one or more TFF cassettes during a first and/or a second tangential flow filtration process, (3) one or more mixing vessels containing a mix of lipids and RNA solution, and/or (4) a lipid- ethanol mixing vessel during an LNP formation process. [0026] In some embodiments, the thermal fluid includes a water-glycol mixture and/or a water- ethylene-glycol mixture, the thermal fluid comprises a pH in a range from about 6.0 to about 8.5, and the density of the thermal fluid does not exceed 1kg/dm^3. [0027] In some embodiments, the thermal fluid includes a water-glycol mixture, and cooling and heating of the heating and cooling jacket is limited to a temperature range from about 2 deg C to about 35 deg C. [0028] In some embodiments, the heating and cooling jacket disposed around the vessel accommodates a vessel volume of at least one of 1 L, 2 L, 5 L, 10 L, 20 L, and 50 L. [0029] In some embodiments, the vessel includes a volume of at least one of 1 L, 2 L, 5 L, 10 L, 20 L, and 50 L. [0030] In some embodiments, each channel of the plurality of channels includes a nominal diameter in a range from about 8 mm to about 20 mm. [0031] In some embodiments, the heating and cooling jacket includes a plurality of vertical spaces vertically separating each channel of the plurality of channels from a channel immediately above and/or below each channel. [0032] In some embodiments, each vertical space of the plurality of vertical spaces includes a height of from about 7 mm to about 10 mm. [0033] In some embodiments: the heating and cooling jacket includes a substantially cylindrical shape; each channel of the plurality of channels extends circumferentially within the heating and cooling jacket 11890709v1 3
Attorney Docket No.: 2013237-0929 around at least a portion of the circumference of the heating and cooling jacket; and each channel of the plurality of channels is disposed above and/or below one or more adjacent channels of the plurality of channels. [0034] In some embodiments, the system and/or jacket includes at least one continuous fluid path through the heating and cooling jacket, the continuous fluid path including each channel of the plurality of channels, wherein the plurality of channels comprises from about 5 to about 20 channels. [0035] In some embodiments, the at least one continuous fluid path includes a plurality of connections disposed at circumferential end(s) of each channel of the plurality of channels, wherein each connection of the plurality of connections connects a channel of the plurality of channels to at least one other channel (i.e., an adjacent channel) of the plurality of channels. [0036] In some embodiments, each connection of the plurality of connections causes the continuous fluid path to make a 180-degree turn. [0037] In some embodiments: the heating and cooling jacket includes a first semi-cylindrical shell and a second semi-cylindrical shell, the continuous fluid path includes at least two continuous fluid paths disposed through the heating and cooling jacket, and the two continuous fluid paths include a first continuous fluid path disposed through the first semi-cylindrical shell and a second continuous fluid path disposed through the second semi-cylindrical shell. [0038] In some embodiments, the heating and cooling jacket includes a height in a range from about 100 mm to about 600 mm. [0039] In some embodiments, the heating and cooling jacket includes a height in a range from about 230 mm to about 300 mm. [0040] In some embodiments, the heating and cooling jacket includes an outer diameter in a range from about 100 mm to about 500 mm. [0041] In some embodiments, the heating and cooling jacket includes an outer diameter in a range from about 230 mm to about 330 mm. [0042] In some embodiments, the heating and cooling jacket includes a height in a range from about 290 mm to about 390 mm. [0043] In some embodiments, each channel of the plurality of channels includes a nominal diameter in a range from about 12 mm to about 15 mm. [0044] In some embodiments, the heating and cooling jacket includes material forming the walls of each channel disposed within each vertical space of the plurality of vertical spaces. [0045] In some embodiments, the heating and cooling jacket includes a channel diameter to channel spacing ratio in a range from about 1.2 to about 2.5. [0046] In some embodiments, the heating and cooling jacket includes a channel diameter to channel spacing ratio in a range from about 1.4 to about 1.9. 11890709v1 4
Attorney Docket No.: 2013237-0929 [0047] In another aspect, the present disclosure is directed to a mobile bioreactor system including: a movable cart; a bioreactor disposed on the moveable cart; and the jacket according to the present disclosure, where the jacket is disposed around the bioreactor. [0048] In some embodiments, the jacket includes a first shell hingedly connected to a second shell, and one of the first shell and the second shell is rigidly coupled to a top surface of the movable cart. [0049] In some embodiments, the movable cart includes wheels and castors that are removably coupled to the cart. [0050] In some embodiments, the bioreactor includes a nominal volume of 50 L. [0051] In some embodiments, the movable cart includes a vertically oriented frame for supporting a bracket, a filter holder, a support plate, and/or an electrical switchbox. [0052] In some embodiments, the jacket comprises channels with an aspect ratio (height to width ratio) of greater than 2.0. [0053] In some embodiments, technologies described herein can be useful for manufacturing RNA-LNP compositions for treatment and/or prevention of coronavirus infection, e.g., SARS-CoV-2 infection, as described in Walsh et al. “RNA-based COVID-19 vaccine BNT162b2 selected for a pivotal efficacy study” medRxiv preprint (2020), which is online accessible at: https://doi.org/10.1101/2020.08.17.20176651; and Milligan et al. “Phase I/II study of COVID-19 RNA vaccine BNT162b1 in adults” Nature (2020 August), which is online accessible at: https://doi.org/10.1038/s41586-020-2639-4, the contents of each of which are incorporated by reference in their entirety. Brief Description of the Drawing [0054] Figure 1 depicts a schematic of an exemplary drug manufacturing process. [0055] Figure 2 depicts an overview of an automated drug manufacturing process, according to aspects of the present disclosure. [0056] Figure 3A illustrates a perspective view of single use bottles in a heating and cooling jacket, according to aspects of the present disclosure. [0057] Figure 3B illustrates a perspective view of single use bottles in a heating and cooling jacket, according to aspects of the present disclosure. [0058] Figure 4 illustrates a support table, according to aspects of the present disclosure. [0059] Figure 5A illustrates details of a single use bottle in a heating and cooling jacket, according to aspects of the present disclosure. [0060] Figure 5B illustrates interior details of a heating and cooling jacket, according to aspects of the present disclosure. 11890709v1 5
Attorney Docket No.: 2013237-0929 [0061] Figure 6A illustrates exterior details of a heating and cooling jacket, according to aspects of the present disclosure. [0062] Figure 6B illustrates interior details of a heating and cooling jacket, according to aspects of the present disclosure. [0063] Figure 7A illustrates further details of a single use bottle in a heating and cooling jacket, according to aspects of the present disclosure. [0064] Figure 7B illustrates a side perspective view of a heating and cooling jacket, according to aspects of the present disclosure. [0065] Figure 7C illustrates details of a heating and cooling jacket sensor access port, according to aspects of the present disclosure. [0066] Figure 8A illustrates an image of exterior details of a heating and cooling jacket, according to aspects of the present disclosure. [0067] Figure 8B illustrates an image of exterior details of a heating and cooling jacket, according to aspects of the present disclosure. [0068] Figure 9A illustrates a perspective view of single use bottle in a heating and cooling jacket, according to aspects of the present disclosure. [0069] Figure 9B illustrates a perspective view of single use bottle in a heating and cooling jacket, according to aspects of the present disclosure. [0070] Figure 10 illustrates a system for heating and cooling single use bottles, according to aspects of the present disclosure. [0071] Figure 11 depicts an overview of an exemplary manufacturing process for a pharmaceutical- grade composition comprising RNA, according to aspects of the present disclosure. [0072] Figure 12 illustrates an overview of exemplary DNA template manufacture process via a PCR-based process, according to aspects of the present disclosure. [0073] Figure 13 illustrates an exemplary process for manufacturing LNP compositions, according to aspects of the present disclosure. [0074] Figure 14 illustrates a method or process for heating or cooling a chemical or biological process, according to aspects of the present disclosure. [0075] Figure 15 illustrates a heating and cooling jacket in an open position, according to aspects of the present disclosure. [0076] Figure 16 illustrates a heating and cooling jacket in a closed position, according to aspects of the present disclosure. [0077] Figure 17 illustrates a heating and cooling jacket in operation, according to aspects of the present disclosure. 11890709v1 6
Attorney Docket No.: 2013237-0929 [0078] Figure 18 illustrates a heating and cooling jacket in a closed position without outer casings, according to aspects of the present disclosure. [0079] Figure 19 illustrates an example of a 1 L heating and cooling jacket, according to aspects of the present disclosure. [0080] Figure 20 illustrates an example of a 5 L heating and cooling jacket, according to aspects of the present disclosure. [0081] Figure 21 illustrates an example of a 10 L heating and cooling jacket, according to aspects of the present disclosure. [0082] Figure 22 illustrates an example of a 20 L heating and cooling jacket, according to aspects of the present disclosure. [0083] Figure 23 illustrates an example of grip features on a 1 L single use bottle in a heating and cooling jacket, according to aspects of the present disclosure. [0084] Figure 24 illustrates an example of grip concavities in a heating and cooling jacket, according to aspects of the present disclosure. [0085] Figure 25 illustrates an interior view of a 50 L heating and cooling jacket, according to aspects of the present disclosure. [0086] Figure 26 illustrates a cross-sectional view of a 50 L heating and cooling jacket, according to aspects of the present disclosure. [0087] Figure 27 illustrates a perspective view of a 50 L heating and cooling jacket, according to aspects of the present disclosure. [0088] Figure 28 illustrates a front view of a mobile bioreactor unit with a heating and cooling jacket, according to aspects of the present disclosure. [0089] Figure 29 illustrates a side view of a mobile bioreactor unit with a heating and cooling jacket, according to aspects of the present disclosure. [0090] Figure 30 illustrates a rear view of a mobile bioreactor unit with a heating and cooling jacket, according to aspects of the present disclosure. [0091] Figure 31 illustrates a front view of a mobile bioreactor unit with a heating and cooling jacket, according to aspects of the present disclosure. [0092] Figure 32 illustrates a perspective view of a mobile bioreactor unit without a heating and cooling jacket, according to aspects of the present disclosure. [0093] Figure 33 illustrates a partial front view of a mobile bioreactor unit with a heating and cooling jacket on load cells, according to aspects of the present disclosure. [0094] Figure 34 illustrates a partial front view of a mobile bioreactor unit with a heating and cooling jacket on a scale, according to aspects of the present disclosure. 11890709v1 7
Attorney Docket No.: 2013237-0929 Certain Definitions [0095] About or Approximately: The term “about” or “approximately”, when used herein in reference to a value, refers to a value that is similar, in context to a stated reference value. In general, those skilled in the art, familiar with the context, will appreciate the relevant degree of variance encompassed by “about” or “approximately” in that context. For example, in some embodiments, the term “about” or “approximately” may encompass a range of values that are within 25%, 20%, 19%, 18%, 17%, 16%, 15%, 14%, 13%, 12%, 11%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, or less of the referred value. [0096] Administration: As used herein, the term “administration” typically refers to the administration of a composition to a subject or system. Those of ordinary skill in the art will be aware of a variety of routes that may, in appropriate circumstances, be utilized for administration to a subject, for example a human. For example, in some embodiments, administration may be ocular, oral, parenteral, topical, etc. In some particular embodiments, administration may be bronchial (e.g., by bronchial instillation), buccal, dermal (which may be or comprise, for example, one or more of topical to the dermis, intradermal, intradermal, transdermal, etc.), enteral, intra-arterial, intradermal, intragastric, intramedullary, intramuscular, intranasal, intraperitoneal, intrathecal, intravenous, intraventricular, within a specific organ (e. g. intrahepatic), mucosal, nasal, oral, rectal, subcutaneous, sublingual, topical, tracheal (e.g., by intratracheal instillation), vaginal, vitreal, etc. In some embodiments, administration may be intramuscular. In some embodiments, administration may involve dosing that is intermittent (e.g., a plurality of doses separated in time) and/or periodic (e.g., individual doses separated by a common period of time) dosing. In some embodiments, administration may involve continuous dosing (e.g., perfusion) for at least a selected period of time. [0097] Agent: In general, the term “agent”, as used herein, is used to refer to an entity (e.g., for example, a lipid, metal, nucleic acid, polypeptide, polysaccharide, small molecule, etc., or complex, combination, mixture or system [e.g., cell, tissue, organism] thereof), or phenomenon (e.g., heat, electric current or field, magnetic force or field, etc.). In appropriate circumstances, as will be clear from context to those skilled in the art, the term may be utilized to refer to an entity that is or comprises a cell or organism, or a fraction, extract, or component thereof. Alternatively or additionally, as context will make clear, the term may be used to refer to a natural product in that it is found in and/or is obtained from nature. In some instances, again as will be clear from context, the term may be used to refer to one or more entities that is man-made in that it is designed, engineered, and/or produced through action of the hand of man and/or is not found in nature. In some embodiments, an agent may be utilized in isolated or pure form; in some embodiments, an agent may be utilized in crude form. In some embodiments, potential agents may be provided as collections or libraries, for example that may be screened to identify or characterize active agents within them. In some cases, the term “agent” may refer to a compound or entity that is or comprises a polymer; in some cases, the term may refer to a compound or entity that comprises one or more polymeric moieties. In some embodiments, the term “agent” may refer to a compound or entity that is not a polymer and/or is substantially free of any polymer and/or of one or more particular polymeric moieties. In some embodiments, the term may refer to a compound or entity that lacks or is substantially free of any polymeric moiety. [0098] Analog: As used herein, the term “analog” refers to a substance that shares one or more particular structural features, elements, components, or moieties with a reference substance. Typically, an 11890709v1 8
Attorney Docket No.: 2013237-0929 “analog” shows significant structural similarity with the reference substance, for example sharing a core or consensus structure, but also differs in certain discrete ways. In some embodiments, an analog is a substance that can be generated from the reference substance, e.g., by chemical manipulation of the reference substance. In some embodiments, an analog is a substance that can be generated through performance of a synthetic process substantially similar to (e.g., sharing a plurality of steps with) one that generates the reference substance. In some embodiments, an analog is or can be generated through performance of a synthetic process different from that used to generate the reference substance. [0099] Antibody agent: As used herein, the term "antibody agent" refers to an agent that specifically binds to a particular antigen. In some embodiments, the term encompasses any polypeptide or polypeptide complex that includes immunoglobulin structural elements sufficient to confer specific binding. Exemplary antibody agents include, but are not limited to monoclonal antibodies or polyclonal antibodies. In some embodiments, an antibody agent may include one or more constant region sequences that are characteristic of mouse, rabbit, primate, or human antibodies. In some embodiments, an antibody agent may include one or more sequence elements are humanized, primatized, chimeric, etc., as is known in the art. In many embodiments, the term "antibody agent" is used to refer to one or more of the art-known or developed constructs or formats for utilizing antibody structural and functional features in alternative presentation. For example, embodiments, an antibody agent utilized in accordance with the present disclosure is in a format selected from, but not limited to, intact IgA, IgG, IgE or IgM antibodies; bi- or multi- specific antibodies (e.g., Zybodies®, etc.); antibody fragments such as Fab fragments, Fab' fragments, F(ab')2 fragments, Fd' fragments, Fd fragments, and isolated complementarity determining regions (CDRs) or sets thereof; single chain Fvs; polypeptide-Fc fusions; single domain antibodies (e.g., shark single domain antibodies such as IgNAR or fragments thereof); cameloid antibodies; masked antibodies (e.g., Probodies®); Small Modular ImmunoPharmaceuticals ("SMIPsTM"); single chain or Tandem diabodies (TandAb®); VHHs; Anticalins®; Nanobodies® minibodies; BiTE®s; ankyrin repeat proteins or DARPINs®; Avimers®; DARTs; TCR-like antibodies; Adnectins®; Affilins®; Trans-bodies®; Affibodies®; TrimerX®; MicroProteins; Fynomers®, Centyrins®; and KALBITOR®s. In some embodiments, an antibody may lack a covalent modification (e.g., attachment of a glycan) that it would have if produced naturally. In some embodiments, an antibody may contain a covalent modification (e.g., attachment of a glycan, a payload [e.g., a detectable moiety, a therapeutic moiety, a catalytic moiety, etc.], or other pendant group [e.g., poly-ethylene glycol, etc.]. In many embodiments, an antibody agent is or comprises a polypeptide whose amino acid sequence includes one or more structural elements recognized by those skilled in the art as a complementarity determining region (CDR); in some embodiments an antibody agent is or comprises a polypeptide whose amino acid sequence includes at least one CDR (e.g., at least one heavy chain CDR and/or at least one light chain CDR) that is substantially identical to one found in a reference antibody. In some embodiments an included CDR is substantially identical to a reference CDR in that it is either identical in sequence or contains between 1-5 amino acid substitutions as compared with the reference CDR. In some embodiments an included CDR is substantially identical to a reference CDR in that it shows at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity with the reference CDR. In some embodiments, an included CDR is substantially identical to a reference CDR in that it shows at least 95%, 96%, 97%, 98%, 99%, or 100% sequence identity with the reference CDR. In some embodiments an included CDR is substantially identical to a reference CDR in that at least one amino acid within the included CDR is deleted, added, or substituted as compared with the reference CDR but the included CDR has an amino acid sequence that is otherwise identical 11890709v1 9
Attorney Docket No.: 2013237-0929 with that of the reference CDR. In some embodiments an included CDR is substantially identical to a reference CDR in that 1-5 amino acids within the included CDR are deleted, added, or substituted as compared with the reference CDR but the included CDR has an amino acid sequence that is otherwise identical to the reference CDR. In some embodiments, an included CDR is substantially identical to a reference CDR in that at least one amino acid within the included CDR is substituted as compared with the reference CDR but the included CDR has an amino acid sequence that is otherwise identical with that of the reference CDR. In some embodiments, an included CDR is substantially identical to a reference CDR in that 1-5 amino acids within the included CDR are deleted, added, or substituted as compared with the reference CDR but the included CDR has an amino acid sequence that is otherwise identical to the reference CDR. In some embodiments, an antibody agent is or comprises a polypeptide whose amino acid sequence includes structural elements recognized by those skilled in the art as an immunoglobulin variable domain. In some embodiments, an antibody agent is a polypeptide protein having a binding domain which is homologous or largely homologous to an immunoglobulin-binding domain. [0100] Antibody agents can be made by the skilled person using methods and commercially available services and kits known in the art. For example, methods of preparation of monoclonal antibodies are well known in the art and include hybridoma technology and phage display technology. Further antibodies suitable for use in the present disclosure are described, for example, in the following publications: Antibodies A Laboratory Manual, Second edition. Edward A. Greenfield. Cold Spring Harbor Laboratory Press (September 30, 2013); Making and Using Antibodies: A Practical Handbook, Second Edition. Eds. Gary C. Howard and Matthew R. Kaser. CRC Press (July 29, 2013); Antibody Engineering: Methods and Protocols, Second Edition (Methods in Molecular Biology). Patrick Chames. Humana Press (August 21, 2012); Monoclonal Antibodies: Methods and Protocols (Methods in Molecular Biology). Eds. Vincent Ossipow and Nicolas Fischer. Humana Press (February 12, 2014); and Human Monoclonal Antibodies: Methods and Protocols (Methods in Molecular Biology). Michael Steinitz. Humana Press (September 30, 2013). [0101] Antibodies may be produced by standard techniques, for example by immunization with the appropriate polypeptide or portion(s) thereof, or by using a phage display library. If polyclonal antibodies are desired, a selected mammal (e.g., mouse, rabbit, goat, horse, etc.) is immunized with an immunogenic polypeptide bearing a desired epitope(s), optionally haptenized to another polypeptide. Depending on the host species, various adjuvants may be used to increase immunological response. Such adjuvants include, but are not limited to, Freund's, mineral gels such as aluminum hydroxide, and surface-active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanin, and dinitrophenol. Serum from the immunized animal is collected and treated according to known procedures. If serum containing polyclonal antibodies to the desired epitope contains antibodies to other antigens, the polyclonal antibodies can be purified by immunoaffinity chromatography or any other method known in the art. Techniques for producing and processing polyclonal antisera are well known in the art. [0102] Antigen: The term “antigen”, as used herein, refers to (i) an agent that elicits an immune response; and/or (ii) an agent that binds to a T cell receptor (e.g., when presented by an MHC molecule) or to an antibody. In some embodiments, an antigen elicits a humoral response (e.g., including production of antigen-specific antibodies); in some embodiments, an antigen elicits a cellular response (e.g., involving T-cells whose receptors specifically interact with the antigen). In some embodiments, an antigen binds to an antibody and may or may not induce a particular physiological response in an organism. In general, an antigen may be 11890709v1 10
Attorney Docket No.: 2013237-0929 or include any chemical entity such as, for example, a small molecule, a nucleic acid, a polypeptide, a carbohydrate, a lipid, a polymer (in some embodiments other than a biologic polymer [e.g., other than a nucleic acid or amino acid polymer]) etc. In some embodiments, an antigen is or comprises a polypeptide. In some embodiments, an antigen is or comprises a glycan. Those of ordinary skill in the art will appreciate that, in general, an antigen may be provided in isolated or pure form, or alternatively may be provided in crude form (e.g., together with other materials, for example in an extract such as a cellular extract or other relatively crude preparation of an antigen-containing source). In some embodiments, antigens utilized in accordance with the present invention are provided in a crude form. In some embodiments, an antigen is a recombinant antigen. [0103] Binding: It will be understood that the term “binding”, as used herein, typically refers to a non-covalent association between or among two or more entities. “Direct” binding involves physical contact between entities or moieties; indirect binding involves physical interaction by way of physical contact with one or more intermediate entities. Binding between two or more entities can typically be assessed in any of a variety of contexts – including where interacting entities or moieties are studied in isolation or in the context of more complex systems (e.g., while covalently or otherwise associated with a carrier entity and/or in a biological system or cell). [0104] Bioreactor: The term “bioreactor” as used herein refers to a vessel used for in vitro transcription described herein. A bioreactor can be of any size so long as it is useful for in vitro transcription. For example, in some embodiments, a bioreactor can be at least 0.5 liter, including 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 liters or more, or any volume in between. The internal conditions of the bioreactor, including, but not limited to pH and temperature, are typically controlled during in vitro transcription. The bioreactor can be composed of any material that is suitable for in vitro transcription under the conditions as described herein, including glass, plastic or metal. One of ordinary skill in the art will be aware of and will be able to choose suitable bioreactor volume for use in practicing in vitro transcription. [0105] Cap: As used herein, the term “cap” refers to a structure comprising or essentially consisting of a nucleoside-5 '-triphosphate that is typically joined to a 5'-end of an uncapped RNA (e.g., an uncapped RNA having a 5'- diphosphate). In some embodiments, a cap is or comprises a guanine nucleotide. In some embodiments, a cap is or comprises a naturally-occurring RNA 5’ cap, including, e.g., but not limited to a N7- methylguanosine cap, which has a structure designated as "m7G." In some embodiments, a cap is or comprises a synthetic cap analog that resembles an RNA cap structure and possesses the ability to stabilize RNA if attached thereto, including, e.g., but not limited to anti-reverse cap analogs (ARCAs) known in the art. Those skilled in the art will appreciate that methods for joining a cap to a 5’ end of an RNA are known in the art. For example, in some embodiments, a capped RNA may be obtained by in vitro capping of RNA that has a 5' triphosphate group or RNA that has a 5' diphosphate group with a capping enzyme system (including, e.g., but not limited to vaccinia capping enzyme system or Saccharomyces cerevisiae capping enzyme system). Alternatively, a capped RNA can be obtained by in vitro transcription (IVT) of a DNA template, wherein, in addition to the GTP, an IVT system also contains a cap analog, e.g., as known in the art. Non-limiting examples of a cap analog include a m7GpppG cap analog or an N7-methyl-, 2’-O- methyl -GpppG ARCA cap analog or an N7-methyl-, 3'-O-methyl-GpppG ARCA cap analog, or any commercially available cap analogs, including, e.g., CleanCap (Trilink), EZ Cap, etc. In some embodiments, a cap analog is or comprises a trinucleotide cap analog. 11890709v1 11
Attorney Docket No.: 2013237-0929 [0106] Comparable: As used herein, the term “comparable” refers to two or more agents, entities, situations, sets of conditions, etc., that may not be identical to one another but that are sufficiently similar to permit comparison there between so that one skilled in the art will appreciate that conclusions may reasonably be drawn based on differences or similarities observed. In some embodiments, comparable sets of conditions, circumstances, individuals, or populations are characterized by a plurality of substantially identical features and one or a small number of varied features. Those of ordinary skill in the art will understand, in context, what degree of identity is required in any given circumstance for two or more such agents, entities, situations, sets of conditions, etc. to be considered comparable. For example, those of ordinary skill in the art will appreciate that sets of circumstances, individuals, or populations are comparable to one another when characterized by a sufficient number and type of substantially identical features to warrant a reasonable conclusion that differences in results obtained or phenomena observed under or with different sets of circumstances, individuals, or populations are caused by or indicative of the variation in those features that are varied. [0107] Complementary: As used herein, the term “complementary” is used in reference to oligonucleotide hybridization related by base-pairing rules. For example, the sequence “C-A-G-T” is complementary to the sequence “G-T-C-A.” Complementarity can be partial or total. Thus, any degree of partial complementarity is intended to be included within the scope of the term “complementary” provided that the partial complementarity permits oligonucleotide hybridization. Partial complementarity is where one or more nucleic acid bases is not matched according to the base pairing rules. Total or complete complementarity between nucleic acids is where each and every nucleic acid base is matched with another base under the base pairing rules. [0108] Detecting: The term “detecting” is used broadly herein to include appropriate means of determining the presence or absence of an entity of interest or any form of measurement of an entity of interest in a sample. Thus, “detecting” may include determining, measuring, assessing, or assaying the presence or absence, level, amount, and/or location of an entity of interest. Quantitative and qualitative determinations, measurements or assessments are included, including semi-quantitative. Such determinations, measurements or assessments may be relative, for example when an entity of interest is being detected relative to a control reference, or absolute. As such, the term “quantifying” when used in the context of quantifying an entity of interest can refer to absolute or to relative quantification. Absolute quantification may be accomplished by correlating a detected level of an entity of interest to known control standards (e.g., through generation of a standard curve). Alternatively, relative quantification can be accomplished by comparison of detected levels or amounts between two or more different entities of interest to provide a relative quantification of each of the two or more different entities of interest, i.e., relative to each other. [0109] Determine: Those of ordinary skill in the art, reading the present specification, will appreciate that a step of “determining” can utilize or be accomplished through use of any of a variety of techniques available to those skilled in the art, including for example specific techniques explicitly referred to herein. In some embodiments, determining involves manipulation of a physical sample. In some embodiments, determining involves consideration and/or manipulation of data or information, for example utilizing a computer or other processing unit adapted to perform a relevant analysis. In some embodiments, determining involves receiving relevant information and/or materials from a source. In some embodiments, determining involves comparing one or more features of a sample or entity to a comparable reference. 11890709v1 12
Attorney Docket No.: 2013237-0929 [0110] Dosage form or unit dosage form: Those skilled in the art will appreciate that the term “dosage form” may be used to refer to a physically discrete unit of an active agent (e.g., a therapeutic or diagnostic agent) for administration to a subject. Typically, each such unit contains a predetermined quantity of active agent. In some embodiments, such quantity is a unit dosage amount (or a whole fraction thereof) appropriate for administration in accordance with a dosing regimen that has been determined to correlate with a desired or beneficial outcome when administered to a relevant population (i.e., with a therapeutic dosing regimen). Those of ordinary skill in the art appreciate that the total amount of a therapeutic composition or agent administered to a particular subject is determined by one or more attending physicians and may involve administration of multiple dosage forms. [0111] Encapsulate: The term “encapsulate” or “encapsulation” is used herein to refer to at least a portion of a component is enclosed or surrounded by another material or another component in a composition. In some embodiments, a component can be fully enclosed or surrounded by another material or another component in a composition. [0112] Excipient: As used herein, the term “excipient” refers to a non-therapeutic agent that may be included in a pharmaceutical composition, for example to provide or contribute to a desired property or effect (e.g., desired consistency, delivery, and/or stabilizing effect, etc.). In some embodiments, suitable pharmaceutical excipients to be added to a LNP composition may include, for example, salts, starch, glucose, lactose, sucrose, gelatin, sodium chloride, glycerol, propylene, glycol, water, ethanol and the like. [0113] Encode: As used herein, the term “encode” or “encoding” refers to sequence information of a first molecule that guides production of a second molecule having a defined sequence of nucleotides (e.g., mRNA) or a defined sequence of amino acids. For example, a DNA molecule can encode an RNA molecule (e.g., by a transcription process that includes a DNA-dependent RNA polymerase enzyme). An RNA molecule can encode a polypeptide (e.g., by a translation process). Thus, a gene, a cDNA, or a single-stranded RNA (e.g., an mRNA) encodes a polypeptide if transcription and translation of mRNA corresponding to that gene produces the polypeptide in a cell or other biological system. In some embodiments, a coding region of a single-stranded RNA encoding a target polypeptide agent refers to a coding strand, the nucleotide sequence of which is identical to the mRNA sequence of such a target polypeptide agent. In some embodiments, a coding region of a single-stranded RNA encoding a target polypeptide agent refers to a non-coding strand of such a target polypeptide agent, which may be used as a template for transcription of a gene or cDNA. [0114] Expression: As used herein, “expression” of a nucleic acid sequence refers to one or more of the following events: (1) production of an RNA template from a DNA sequence (e.g., by transcription); (2) processing of an RNA transcript (e.g., by splicing, editing, 5’ cap formation, and/or 3’ end formation); (3) translation of an RNA into a polypeptide or protein; and/or (4) post-translational modification of a polypeptide or protein. [0115] Fed-batch process: The term “fed-batch process” as used herein refers to a process in which one or more components are introduced into a vessel, e.g., a bioreactor, at some time subsequent to the beginning of a reaction. In some embodiments, one or more components are introduced by a fed-batch process to maintain its concentration low during a reaction. In some embodiments, one or more components are introduced by a fed-batch process to replenish what is depleted during a reaction. 11890709v1 13
Attorney Docket No.: 2013237-0929 [0116] Five prime untranslated region: As used herein, the terms "five prime untranslated region" or "5' UTR" refer to a sequence of an mRNA molecule that begins at the transcription start site and ends one nucleotide (nt) before the start codon (usually AUG) of the coding region of an RNA. [0117] Functional: As used herein, a “functional” biological molecule is a biological molecule in a form in which it exhibits a property and/or activity by which it is characterized. In some embodiments, a biological molecule may have two functions (i.e., bifunctional) or many functions (i.e., multifunctional). [0118] Gene: As used herein, the term “gene” refers to a DNA sequence in a chromosome that codes for a product (e.g., an RNA product and/or a polypeptide product). In some embodiments, a gene includes coding sequence (i.e., sequence that encodes a particular product); in some embodiments, a gene includes non-coding sequence. In some particular embodiments, a gene may include both coding (e.g., exonic) and non-coding (e.g., intronic) sequences. In some embodiments, a gene may include one or more regulatory elements that, for example, may control or impact one or more aspects of gene expression (e.g., cell-type- specific expression, inducible expression, etc.). [0119] Gene product or expression product: As used herein, the term “gene product” or “expression product” generally refers to an RNA transcribed from the gene (pre-and/or post-processing) or a polypeptide (pre- and/or post-modification) encoded by an RNA transcribed from the gene. [0120] Homology: As used herein, the term “homology” or “homolog” refers to the overall relatedness between polynucleotide molecules (e.g., DNA molecules and/or RNA molecules) and/or between polypeptide molecules. In some embodiments, polynucleotide molecules (e.g., DNA molecules and/or RNA molecules) and/or polypeptide molecules are considered to be “homologous” to one another if their sequences are at least 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 99% identical. In some embodiments, polynucleotide molecules (e.g., DNA molecules and/or RNA molecules) and/or polypeptide molecules are considered to be “homologous” to one another if their sequences are at least 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 99% similar (e.g., containing residues with related chemical properties at corresponding positions). For example, as is well known by those of ordinary skill in the art, certain amino acids are typically classified as similar to one another as "hydrophobic" or “hydrophilic” amino acids, and/or as having “polar” or “non-polar” side chains. Substitution of one amino acid for another of the same type may often be considered a “homologous” substitution. [0121] Host cell: As used herein, refers to a cell into which exogenous material (e.g., DNA such as recombinant or otherwise) has been introduced. Persons of skill upon reading this disclosure will understand that such terms refer not only to the particular subject cell, but also to the progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term "host cell" as used herein. In some embodiments, host cells include prokaryotic and eukaryotic cells selected from any of the Kingdoms of life that are suitable for expressing an exogenous DNA (e.g., a recombinant nucleic acid sequence). Exemplary cells include those of prokaryotes and eukaryotes (single-cell or multiple- cell), bacterial cells (e.g., strains of E. coli, Bacillus spp., Streptomyces spp., etc.), mycobacteria cells, fungal cells, yeast cells (e.g., S. cerevisiae, S. pombe, P. pastoris, P. methanolica, etc.), plant cells, insect cells (e.g., SF-9, SF-21, baculovirus-infected insect cells, Trichoplusia ni, etc.), non-human animal cells, human cells, or cell 11890709v1 14
Attorney Docket No.: 2013237-0929 fusions such as, for example, hybridomas or quadromas. In some embodiments, a host cell is a human, monkey, ape, hamster, rat, or mouse cell. In some embodiments, a host cell is eukaryotic. For example, an eukaryotic host cell may be CHO (e.g., CHO Kl, DXB-11 CHO, Veggie-CHO), COS (e.g., COS-7), retinal cell, Vero, CV1, kidney (e.g., HEK293, 293 EBNA, MSR 293, MDCK, HaK, BHK), HeLa, HepG2, WI38, MRC 5, Colo205, HB 8065, HL-60, (e.g., BHK21), Jurkat, Daudi, A431 (epidermal), CV-1, U937, 3T3, L cell, C127 cell, SP2/0, NS-0, MMT 060562, Sertoli cell, BRL 3 A cell, HT1080 cell, myeloma cell, tumor cell, or a cell line derived from an aforementioned cell. [0122] Identity: As used herein, the term “identity” refers to the overall relatedness between polymeric molecules, e.g., between nucleic acid molecules (e.g., DNA molecules and/or RNA molecules) and/or between polypeptide molecules. In some embodiments, polymeric molecules are considered to be “substantially identical” to one another if their sequences are at least 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 99% identical. Calculation of the percent identity of two nucleic acid or polypeptide sequences, for example, can be performed by aligning the two sequences for optimal comparison purposes (e.g., gaps can be introduced in one or both of a first and a second sequences for optimal alignment and non-identical sequences can be disregarded for comparison purposes). In certain embodiments, the length of a sequence aligned for comparison purposes is at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or substantially 100% of the length of a reference sequence. The nucleotides at corresponding positions are then compared. When a position in the first sequence is occupied by the same residue (e.g., nucleotide or amino acid) as the corresponding position in the second sequence, then the molecules are identical at that position. The percent identity between the two sequences is a function of the number of identical positions shared by the sequences, taking into account the number of gaps, and the length of each gap, which needs to be introduced for optimal alignment of the two sequences. The comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm. For example, the percent identity between two nucleotide sequences can be determined using the algorithm of Meyers and Miller (CABIOS, 1989, 4: 11-17), which has been incorporated into the ALIGN program (version 2.0). In some exemplary embodiments, nucleic acid sequence comparisons made with the ALIGN program use a PAM120 weight residue table, a gap length penalty of 12 and a gap penalty of 4. The percent identity between two nucleotide sequences can, alternatively, be determined using the GAP program in the GCG software package using an NWSgapdna.CMP matrix. [0123] Improved, increased or reduced: As used herein, these terms, or grammatically comparable comparative terms, indicate values that are relative to a comparable reference measurement. For example, in some embodiments, an assessed value achieved with an agent of interest may be “improved” relative to that obtained with a comparable reference agent. Alternatively or additionally, in some embodiments, an assessed value achieved in a subject or system of interest may be “improved” relative to that obtained in the same subject or system under different conditions (e.g., prior to or after an event such as administration of an agent of interest), or in a different, comparable subject (e.g., in a comparable subject or system that differs from the subject or system of interest in presence of one or more indicators of a particular disease, disorder or condition of interest, or in prior exposure to a condition or agent, etc.). In some embodiments, comparative terms refer to statistically relevant differences (e.g., that are of a prevalence and/or magnitude sufficient to achieve statistical relevance). Those skilled in the art will be aware, or will readily be 11890709v1 15
Attorney Docket No.: 2013237-0929 able to determine, in a given context, a degree and/or prevalence of difference that is required or sufficient to achieve such statistical significance. [0124] In vitro: The term “in vitro” as used herein refers to events that occur in an artificial environment, e.g., in a test tube or reaction vessel (e.g., a bioreactor), in cell culture, etc., rather than within a multi-cellular organism. [0125] In vitro transcription: As used herein, the term "in vitro transcription" or "IVT" refers to the process whereby transcription occurs in vitro in a non-cellular system to produce a synthetic RNA product for use in various applications, including, e.g., production of protein or polypeptides. Such synthetic RNA products can be translated in vitro or introduced directly into cells, where they can be translated. Such synthetic RNA products include, e.g., but not limited to mRNAs, antisense RNA molecules, shRNA molecules, long non- coding RNA molecules, ribozymes, aptamers, guide RNAs (e.g., for CRISPR), ribosomal RNAs, small nuclear RNAs, small nucleolar RNAs, and the like. An IVT reaction typically utilizes a DNA template (e.g., a linear DNA template) as described and/or utilized herein, ribonucleotides (e.g., non-modified ribonucleotide triphosphates or modified ribonucleotide triphosphates), and an appropriate RNA polymerase. [0126] In vitro transcription RNA composition: As used herein, the term “in vitro transcription RNA composition” refers to a composition comprising target RNA synthesized by in vitro transcription. In some embodiments, such a composition can comprise excess in vitro transcription reagents (including, e.g., ribonucleotides and/or capping agents), nucleic acids or fragments thereof such as DNA templates or fragments thereof, polypeptides or fragments thereof such as recombinant enzymes or host cell proteins or fragments thereof, and/or other impurities. In some embodiments, an in vitro transcription RNA composition may have been treated and/or processed prior to a purification process that ultimately produces an RNA transcript preparation comprising RNA transcript at a desired concentration in an appropriate buffer for formulation and/or further manufacturing and/or processing. For example, in some embodiments, an in vitro transcription RNA composition may have been treated to remove or digest DNA template (e.g., using a DNase). In some embodiments, an in vitro transcription RNA composition may have been treated to remove or digest polypeptides (e.g., enzymes such as RNA polymerases, RNase inhibitors, etc.) present in an in vitro transcription reaction (e.g., using a protease). [0127] In vivo: As used herein, the term “in vivo” refers to events that occur within a multi-cellular organism, such as a human and a non-human animal. [0128] Nanoparticle: As used herein, the term “nanoparticle” refers to a particle having a diameter of less than 1000 nanometers (nm). In some embodiments, a nanoparticle has a diameter of less than 300 nm, as defined by the National Science Foundation. In some embodiments, a nanoparticle has a diameter of less than 100 nm as defined by the National Institutes of Health. In some embodiments, a nanoparticle has a diameter of less than 80 nm as defined by the National Institutes of Health. In some embodiments, a nanoparticle comprises one or more enclosed compartments, separated from the bulk solution by a membrane, which surrounds and encloses a space or compartment. [0129] Nucleic acid/ Polynucleotide: As used herein, the term “nucleic acid” refers to a polymer of at least 2 nucleotides or more, including, e.g., at least 3 nucleotides, at least 4 nucleotides, at least 5 nucleotides, at least 6 nucleotides, at least 7 nucleotides, at least 8 nucleotides, at least 9 nucleotides, at least 11890709v1 16
Attorney Docket No.: 2013237-0929 10 nucleotides, or more. In some embodiments, a nucleic acid is or comprises DNA. In some embodiments, a nucleic acid is or comprises RNA. In some embodiments, a nucleic acid is or comprises peptide nucleic acid (PNA). In some embodiments, a nucleic acid is or comprises a single stranded nucleic acid. In some embodiments, a nucleic acid is or comprises a double-stranded nucleic acid. In some embodiments, a nucleic acid comprises both single and double-stranded portions. In some embodiments, a nucleic acid comprises a backbone that comprises one or more phosphodiester linkages. In some embodiments, a nucleic acid comprises a backbone that comprises both phosphodiester and non-phosphodiester linkages. For example, in some embodiments, a nucleic acid may comprise a backbone that comprises one or more phosphorothioate or 5'-N-phosphoramidite linkages and/or one or more peptide bonds, e.g., as in a “peptide nucleic acid”. In some embodiments, a nucleic acid comprises one or more, or all, natural residues (e.g., adenine, cytosine, deoxyadenosine, deoxycytidine, deoxyguanosine, deoxythymidine, guanine, thymine, uracil). In some embodiments, a nucleic acid comprises on or more, or all, non-natural residues. In some embodiments, a non- natural residue comprises a nucleoside analog (e.g., 2-aminoadenosine, 2-thiothymidine, inosine, pyrrolo- pyrimidine, 3 -methyl adenosine, 5-methylcytidine, C-5 propynyl-cytidine, 1-methyl-pseudouridine, C-5 propynyl-uridine, 2-aminoadenosine, C5-bromouridine, C5-fluorouridine, C5-iodouridine, C5-propynyl-uridine, C5 -propynyl-cytidine, C5-methylcytidine, 2-aminoadenosine, 7-deazaadenosine, 7-deazaguanosine, 8- oxoadenosine, 8-oxoguanosine, 6-O-methylguanine, 2-thiocytidine, methylated bases, intercalated bases, and combinations thereof). In some embodiments, a non-natural residue comprises one or more modified sugars (e.g., 2'-fluororibose, ribose, 2'-deoxyribose, arabinose, and hexose) as compared to those in natural residues. In some embodiments, a nucleic acid has a nucleotide sequence that encodes a functional gene product such as an RNA or polypeptide. In some embodiments, a nucleic acid has a nucleotide sequence that comprises one or more introns. In some embodiments, a nucleic acid may be prepared by isolation from a natural source, enzymatic synthesis (e.g., by polymerization based on a complementary template, e.g., in vivo or in vitro, reproduction in a recombinant cell or system, or chemical synthesis. In some embodiments, a nucleic acid is at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 225, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475, 500, 600, 700, 800, 900, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 5500, 6000, 6500, 7000, 7500, 8000, 8500, 9000, 9500, 10,000, 10,500, 11,000, 11,500, 12,000, 12,500, 13,000, 13,500, 14,000, 14,500, 15,000, 15,500, 16,000, 16,500, 17,000, 17,500, 18,000, 18,500, 19,000, 19,500, or 20,000 or more residues or nucleotides long. [0130] Pharmaceutical grade: The term “pharmaceutical grade” as used herein refers to standards for chemical and biological drug substances, drug products, dosage forms, compounded preparations, excipients, medical devices, and dietary supplements, established by a recognized national or regional pharmacopeia (e.g., The United States Pharmacopeia and The Formulary (USP–NF)). [0131] Polypeptide: The term “polypeptide”, as used herein, typically has its art-recognized meaning of a polymer of at least three amino acids or more. Those of ordinary skill in the art will appreciate that the term “polypeptide” is intended to be sufficiently general as to encompass not only polypeptides having a complete sequence recited herein, but also to encompass polypeptides that represent functional, biologically active, or characteristic fragments, portions or domains (e.g., fragments, portions, or domains retaining at least one activity) of such complete polypeptides. In some embodiments, polypeptides may contain L-amino acids, D-amino acids, or both and/or may contain any of a variety of amino acid modifications or analogs known in the 11890709v1 17
Attorney Docket No.: 2013237-0929 art. Useful modifications include, e.g., terminal acetylation, amidation, methylation, etc. In some embodiments, polypeptides may comprise natural amino acids, non-natural amino acids, synthetic amino acids, and combinations thereof (e.g., may be or comprise peptidomimetics). In some embodiments, a polypeptide may be or comprise an enzyme. In some embodiments, a polypeptide may be or comprise a polypeptide antigen. In some embodiments, a polypeptide may be or comprise an antibody agent. In some embodiments a polypeptide may be or comprise a cytokine. [0132] Pure or Purified: As used herein, an agent or entity is “pure” or “purified” if it is substantially free of other components. For example, a preparation that contains more than about 90% of a particular agent or entity is typically considered to be a pure preparation. In some embodiments, an agent or entity is at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% pure in a preparation. [0133] Ribonucleotide: As used herein, the term “ribonucleotide” encompasses unmodified ribonucleotides and modified ribonucleotides. For example, unmodified ribonucleotides include the purine bases adenine (A) and guanine (G), and the pyrimidine bases cytosine (C) and uracil (U). Modified ribonucleotides may include one or more modifications including, but not limited to, for example, (a) end modifications, e.g., 5' end modifications (e.g., phosphorylation, dephosphorylation, conjugation, inverted linkages, etc.), 3' end modifications (e.g., conjugation, inverted linkages, etc.), (b) base modifications, e.g. , replacement with modified bases, stabilizing bases, destabilizing bases, or bases that base pair with an expanded repertoire of partners, or conjugated bases, (c) sugar modifications (e.g., at the 2' position or 4' position) or replacement of the sugar, and (d) internucleoside linkage modifications, including modification or replacement of the phosphodiester linkages. The term “ribonucleotide” also encompasses ribonucleotide triphosphates including modified and non-modified ribonucleotide triphosphates. [0134] Ribonucleic acid (RNA): As used herein, the term “RNA” refers to a polymer of ribonucleotides. In some embodiments, an RNA is single stranded. In some embodiments, an RNA is double stranded. In some embodiments, an RNA comprises both single and double stranded portions. In some embodiments, an RNA can comprise a backbone structure as described in the definition of “Nucleic acid / Polynucleotide” above. An RNA can be a regulatory RNA (e.g., siRNA, microRNA, etc.), or a messenger RNA (mRNA). In some embodiments, an RNA is a mRNA. In some embodiments, where an RNA is a mRNA, an RNA typically comprises at its 3’ end a poly(A) region. In some embodiments where an RNA is a mRNA, an RNA typically comprises at its 5’ end, an art-recognized cap structure, e.g., for recognizing and attachment of a mRNA to a ribosome to initiate translation. In some embodiments, an RNA is a synthetic RNA. Synthetic RNAs include RNAs that are synthesized in vitro (e.g., by enzymatic synthesis methods and/or by chemical synthesis methods). In some embodiments, an RNA is a single-stranded RNA. In some embodiments, a single-stranded RNA may comprise self-complementary elements and/or may establish a secondary and/or tertiary structure. One of ordinary skill in the art will understand that when a single-stranded RNA is referred to as “encoding,” it can mean that it comprises a nucleic acid sequence that itself encodes or that it comprises a complement of the nucleic acid sequence that encodes. In some embodiments, a single-stranded RNA can be a self-amplifying RNA (also known as self-replicating RNA). [0135] Recombinant: as used herein, is intended to refer to polypeptides that are designed, engineered, prepared, expressed, created, manufactured, and/or or isolated by recombinant means, such as 11890709v1 18
Attorney Docket No.: 2013237-0929 polypeptides expressed using a recombinant expression vector transfected into a host cell; polypeptides isolated from a recombinant, combinatorial human polypeptide library; polypeptides isolated from an animal (e.g., a mouse, rabbit, sheep, fish, etc.) that is transgenic for or otherwise has been manipulated to express a gene or genes, or gene components that encode and/or direct expression of the polypeptide or one or more component(s), portion(s), element(s), or domain(s) thereof; and/or polypeptides prepared, expressed, created or isolated by any other means that involves splicing or ligating selected nucleic acid sequence elements to one another, chemically synthesizing selected sequence elements, and/or otherwise generating a nucleic acid that encodes and/or directs expression of the polypeptide or one or more component(s), portion(s), element(s), or domain(s) thereof. In some embodiments, one or more of such selected sequence elements is found in nature. In some embodiments, one or more of such selected sequence elements is designed in silico. In some embodiments, one or more such selected sequence elements results from mutagenesis (e.g., in vivo or in vitro) of a known sequence element, e.g., from a natural or synthetic source such as, for example, in the germline of a source organism of interest (e.g., of a human, a mouse, etc.). [0136] Reference: As used herein, the term “reference” describes a standard or control relative to which a comparison is performed. For example, in some embodiments, an agent, animal, individual, population, sample, sequence or value of interest is compared with a reference or control agent, animal, individual, population, sample, sequence or value. In some embodiments, a reference or control is tested and/or determined substantially simultaneously with the testing or determination of interest. In some embodiments, a reference or control is a historical reference or control, optionally embodied in a tangible medium. Typically, as would be understood by those skilled in the art, a reference or control is determined or characterized under comparable conditions or circumstances to those under assessment. Those skilled in the art will appreciate when sufficient similarities are present to justify reliance on and/or comparison to a particular possible reference or control. [0137] RNA polymerase: As used herein, the term “RNA polymerase” refers to an enzyme that catalyzes polyribonucleotide synthesis by addition of ribonucleotide units to a nucleotide chain using DNA or RNA as a template. The term refers to either a complete enzyme as it occurs in nature, or an isolated, active catalytic or functional domain, or fragment thereof. In some embodiments, an RNA polymerase enzyme initiates synthesis at the 3'-end of a primer or a nucleic acid strand, or at a promoter sequence, and proceeds in the 5'- direction along the target nucleic acid to synthesize a strand complementary to the target nucleic acid until synthesis terminates. [0138] RNA transcript preparation: The term “RNA transcript preparation” as used herein refers to a preparation comprising RNA transcript that is purified from an in vitro transcription RNA composition described herein. In some embodiments, an RNA transcript preparation is a preparation comprising pharmaceutical-grade RNA transcript. In some embodiments, an RNA transcript preparation is a preparation comprising RNA transcript, which includes one or more product quality attributes that are characterized and determined to meet a release and/or acceptance criteria (e.g., as described herein). Examples of such product quality attributes include, but are not limited to appearance, RNA length, identity of drug substance as RNA, RNA integrity, RNA sequence, RNA concentration, pH, osmolality, residual DNA template, residual double stranded RNA, bacterial endotoxins, bioburden, and combinations thereof. 11890709v1 19
Attorney Docket No.: 2013237-0929 [0139] Room temperature: As used herein, the term “room temperature” refers to an ambient temperature. In some embodiments, a room temperature is about 18 deg C-30 deg C, e.g., about 18 deg C -25 deg C, or about 20 deg C -25 deg C, or about 20 deg C -30 deg C, or about 23 deg C -27 deg C or about 25 deg C. [0140] Sample: As used herein, the term “sample” typically refers to an aliquot of material obtained or derived from a source of interest, e.g., as described herein. In some embodiments, a source of interest is a biological or environmental source. In some embodiments, a source of interest may be or comprise a cell or an organism, such as a microbe, a plant, or an animal (e.g., a mouse). In some embodiments, a source of interest is or comprises biological tissue or fluid. In some embodiments, a biological fluid may be or comprise an intracellular fluid, an extracellular fluid, an intravascular fluid (blood plasma), an interstitial fluid, a lymphatic fluid, and/or a transcellular fluid. In some embodiments, a biological tissue or sample may be obtained, for example, by aspirate, biopsy (e.g., fine needle or tissue biopsy), swab (e.g., oral, nasal, skin, or vaginal swab), scraping, surgery, washing or lavage (e.g., bronchioalveolar, ductal, nasal, ocular, oral, uterine, vaginal, or other washing or lavage). In some embodiments, a sample is or comprises cells obtained from a subject. In some embodiments, a sample is a “primary sample” obtained directly from a source of interest by any appropriate means. In some embodiments, as will be clear from context, the term “sample” refers to a preparation that is obtained by processing (e.g., by removing one or more components of and/or by adding one or more agents to) a primary sample. For example, a “processed sample” may comprise, for example nucleic acids or proteins extracted from a sample or obtained by subjecting a primary sample to one or more techniques such as amplification or reverse transcription of nucleic acid, isolation and/or purification of certain components, etc. [0141] Stable: The term “stable,” when applied to nucleic acids and/or compositions comprising nucleic acids, e.g., encapsulated in lipid nanoparticles, means that such nucleic acids and/or compositions maintain one or more aspects of their characteristics (e.g., physical and/or structural characteristics, function, and/or activity) over a period of time under a designated set of conditions (e.g., pH, temperature, light, relative humidity, etc.). In some embodiments, such stability is maintained over a period of time of at least about one hour; in some embodiments, such stability is maintained over a period of time of about 5 hours, about 10 hours, about one (1) day, about one (1) week, about two (2) weeks, about one (1) month, about two (2) months, about three (3) months, about four (4) months, about five (5) months, about six (6) months, about eight (8) months, about ten (10) months, about twelve (12) months, about twenty-four (24) months, about thirty-six (36) months, or longer. In some embodiments, such stability is maintained over a period of time within the range of about one (1) day to about twenty-four (24) months, about two (2) weeks to about twelve (12) months, about two (2) months to about five (5) months, etc. In some embodiments, such stability is maintained under an ambient condition (e.g., at room temperature and ambient pressure). In some embodiments, such stability is maintained under a physiological condition (e.g., in vivo or at about 37 ºC for example in serum or in phosphate buffered saline). In some embodiments, such stability is maintained under cold storage (e.g., at or below about 4 ºC, including, e.g., -20 ºC, or -70 ºC). In some embodiments, such stability is maintained when nucleic acids and/or compositions comprising the same are protected from light (e.g., maintaining in the dark). [0142] As an example, in some embodiments, the term “stable” is used in reference to a nanoparticle composition (e.g., a lipid nanoparticle composition). In such embodiments, a stable nanoparticle 11890709v1 20
Attorney Docket No.: 2013237-0929 composition (e.g., a stable nanoparticle composition) and/or component(s) thereof maintain one or more aspects of its characteristics (e.g., physical and/or structural characteristics, function(s), and/or activity) over a period of time under a designated set of conditions. For example, in some embodiments, a stable nanoparticle composition (e.g., a lipid nanoparticle composition) is characterized in that average particle size, particle size distribution, and/or polydispersity of nanoparticles is substantially maintained (e.g., within 10% or less, as compared to the initial characteristic(s)) over a period of time (e.g., as described herein) under a designated set of conditions (e.g., as described herein). In some embodiments, a stable nanoparticle composition (e.g., a lipid nanoparticle composition) is characterized in that no detectable amount of degradation products (e.g., associated with hydrolysis and/or enzymatic digestion) is present after it is maintained under a designated set of conditions (e.g., as described herein) over a period of time. [0143] Synthetic: As used herein, the term “synthetic” refers to an entity that is artificial, or that is made with human intervention, or that results from synthesis rather than naturally occurring. For example, in some embodiments, a synthetic nucleic acid or polynucleotide refers to a nucleic acid molecule that is chemically synthesized, e.g., in some embodiments by solid-phase synthesis. In some embodiments, the term “synthetic” refers to an entity that is made outside of biological cells. For example, in some embodiments, a synthetic nucleic acid or polynucleotide refers to a nucleic acid molecule (e.g., an RNA) that is produced by in vitro transcription using a template. [0144] Tangential flow filtration (TFF): As used herein, the term “tangential flow filtration (TFF)” refers to a mode of operation of a filtration system where a fluid passes parallel to a filter membrane (i.e., the flow is tangential to the membrane) to reduce clogging of the filter. In some embodiments, particles larger than the pore size will flow past the membrane as retentate and is recycled back to the feed. In some embodiments, particles smaller than the pore size will pass through and be collected as permeate. [0145] Three prime untranslated region: As used herein, the terms "three prime untranslated region" or "3' UTR" refer to the sequence of an mRNA molecule that begins following the stop codon of the coding region of an open reading frame sequence. In some embodiments, the 3' UTR begins immediately after the stop codon of the coding region of an open reading frame sequence. In other embodiments, the 3' UTR does not begin immediately after stop codon of the coding region of an open reading frame sequence. [0146] Threshold level (e.g., acceptance criteria): As used herein, the term “threshold level” refers to a level that are used as a reference to attain information on and/or classify the results of a measurement, for example, the results of a measurement attained in an assay. For example, in some embodiments, a threshold level means a value measured in an assay that defines the dividing line between two subsets of a population (e.g., a batch that satisfy quality control criteria vs. a batch that does not satisfy quality control criteria). Thus, a value that is equal to or higher than the threshold level defines one subset of the population, and a value that is lower than the threshold level defines the other subset of the population. A threshold level can be determined based on one or more control samples or across a population of control samples. A threshold level can be determined prior to, concurrently with, or after the measurement of interest is taken. In some embodiments, a threshold level can be a range of values. [0147] Vector: As used herein, the term “vector” refers to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked. One type of vector is a "plasmid", which refers to a circular double stranded DNA into which additional DNA segments may be ligated. Another type of vector is a 11890709v1 21
Attorney Docket No.: 2013237-0929 viral vector, wherein additional DNA segments may be ligated into the viral genome. Certain vectors are capable of autonomous replication in a host cell into which they are introduced (e.g., bacterial vectors having a bacterial origin of replication and episomal mammalian vectors). Other vectors (e.g., non-episomal mammalian vectors) can be integrated into the genome of a host cell upon introduction into the host cell, and thereby are replicated along with the host genome. Moreover, certain vectors are capable of directing the expression of genes to which they are operatively linked. Such vectors are referred to herein as "expression vectors." [0148] Standard techniques may be used for recombinant DNA, oligonucleotide synthesis, and tissue culture and transformation (e.g., electroporation, lipofection). Enzymatic reactions and purification techniques may be performed according to manufacturer's specifications or as commonly accomplished in the art or as described herein. The foregoing techniques and procedures may be generally performed according to conventional methods well known in the art and as described in various general and more specific references that are cited and discussed throughout the present specification. See e.g., Green and Sambrook, Molecular Cloning: A Laboratory Manual (4th ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (2012)), which is incorporated herein by reference for any purpose. Detailed Description of Certain Embodiments [0149] Nucleic acid therapeutics, and particularly RNA therapeutics represent a particularly promising class of therapies for treatment and prevention of various diseases such as cancer, infectious diseases, and/or diseases or disorders associated with overabundance or deficiency in certain proteins. [0150] RNA therapeutics in particular provide remarkably effective as vaccines to address the COVID19 pandemic. Particularly given the promise of this technology, and its adaptability to a wide variety of clinical contexts, including massively large scale (e.g., vaccination and/or treatment on a global scale such as is under development for SARS-CoV-2), improvements to manufacturing technologies, especially those applicable to large-scale production, are especially valuable. [0151] Development of effective delivery technologies has been central to the success of nucleic acid therapeutics, and lipid nanoparticle technologies have proven to be particularly effective (reviewed in, for example, Cullis et al. Molecular Therapy 25:1467, July 5, 2017; See also, US Patent 8058069), specifically including for RNA therapeutics (reviewed in, for example, Hou et al., Nat. Rev. Mater doi.org/10.1038/s41578- 021-00358-0, August 10, 2021). [0152] Technologies provided herein are useful, among other things, to achieve particularly effective and/or efficient production, e.g., on commercial scale and/or under commercial conditions, of pharmaceutical grade LNP preparations and/or compositions (e.g., nucleic acid-LNP preparations, and specifically RNA-LNP preparations). For example, in various embodiments, provided technologies permit and/or facilitate achievement of requirements unique to pharmaceutical-grade (and/or scale) production such as, for example, batch size and/or rate of production, pre-determined in-process controls and/or lot release specifications (e.g., high purity, integrity, potency, and/or stability, etc.), etc. 11890709v1 22
Attorney Docket No.: 2013237-0929 [0153] The present disclosure provides technologies for manufacturing LNP compositions (e.g., including RNA, e.g., therapeutic RNA such as therapeutic mRNA). In some embodiments, provided technologies are useful for manufacturing pharmaceutical-grade RNA-LNP therapeutics. [0154] In some embodiments, provided technologies are useful for large scale manufacturing of LNP (e.g., nucleic acid-LNP, e.g., RNA-LNP) therapeutics, e.g., pharmaceutical-grade therapeutics. For example, in some such embodiments, technologies provided herein can be used to produce a pharmaceutical-grade batch throughput of at least 10,000 vials of LNP (e.g., nucleic acid-LNP, e.g., RNA-LNP) therapeutics (including, e.g., at least 20,000 vials, at least 30,000 vials, at least 40,000 vials, at least 50,000 vials, at least 60,000 vials, at least 70,000 vials, at least 80,000 vials, at least 90,000 vials, at least 100,000 vials, at least 200,000 vials, at least 300,000 vials, at least 400,000 vials, at least 500,000 vials, or more). For example, in some such embodiments, technologies provided herein can be used to produce a pharmaceutical-grade batch throughput of at least 50 L of LNP (e.g., nucleic acid-LNP, e.g., RNA-LNP) therapeutics (including e.g., at least 50 L, at least 60 L, at least 70 L, at least 80 L, at least 100 L, at least 110 L, at least 120 L, at least 130 L, at least 140 L, at least 150 L or more. In some embodiments, each vial can comprise an RNA drug product in an amount of 0.01 mg to 0.5 mg (e.g., 0.01 mg, 0.02 mg, 0.03 mg, 0.04 mg, 0.05 mg, 0.06 mg, 0.07 mg, 0.08 mg, 0.09 mg, 0.1 mg, 0.15 mg, 0.2 mg, 0.25 mg, 0.3 mg, 0.35 mg, 0.4 mg, 0.45 mg, 0.5 mg). [0155] Technologies described herein can be useful for manufacturing LNP (e.g., nucleic acid-LNP, e.g., RNA-LNP) compositions for treatment and/or prevention of a disease, disorder, or condition (e.g., cancer, infectious diseases, diseases associated with protein deficiency, etc.). In some embodiments, technologies described herein can be useful for manufacturing LNP (e.g., nucleic acid-LNP, e.g., RNA-LNP) compositions that comprise or deliver (e.g., by comprising and/or delivering a nucleic acid, such as an RNA, that encodes it) a polypeptide. [0156] In some particular embodiments, technologies described herein can be useful for manufacturing LNP (e.g., nucleic acid-LNP, e.g., RNA-LNP) compositions for inducing an immune response to an antigen. In some embodiments, technologies described herein can be useful for manufacturing LNP(e.g., nucleic acid-LNP, e.g., RNA-LNP) compositions for treatment and/or prevention of coronavirus infection, e.g., SARS-CoV-2 infection, as described in Walsh et al. “RNA-based COVID-19 vaccine BNT162b2 selected for a pivotal efficacy study” medRxiv preprint (2020), which is online accessible at: https://doi.org/10.1101/2020.08.17.20176651; and Milligan et al. “Phase I/II study of COVID-19 RNA vaccine BNT162b1 in adults” Nature (2020 August), which is online accessible at: https://doi.org/10.1038/s41586-020- 2639-4, the contents of each of which are incorporated by reference in their entirety. Lipid Nanoparticles [0157] Those skilled in the art are aware that lipid nanoparticles have achieved successful clinical delivery of a wide range of therapeutic agents including, for example, small molecules, and various nucleic acids – e.g., oligonucleotides, siRNAs, and mRNAs (reviewed, for example, in Hu et al., Nat. Rev. Mater. https://doi.org/10.1038/s41578-021-00358-0, August 10, 2021). [0158] Various routes of administration for lipid nanoparticle compositions have been proposed and/or tested; those skilled in the art will be aware of appropriate routes for particular compositions (e.g., depending on agent being delivered). To give but a few examples, in some embodiments, LNPs are 11890709v1 23
Attorney Docket No.: 2013237-0929 parenterally administered; most clinical studies have utilized parenteral administration, and particularly intravenous, subcutaneous, intradermal, intravitreal, intratumoral, or intramuscular injection. Intrautero injection has also been described. In some embodiments, topical administration is utilized. In some embodiments, intranasal administration is utilized. [0159] In some embodiments, administered LNPs are delivered to or accumulate in the liver. Given that the liver is naturally effective at producing and secreting proteins, liver delivery can prove useful for achieving delivery of an LNP-encapsulated agent (and/or, in the case of a nucleic acid agent such as an RNA agent, a polypeptide encoded thereby) into the bloodstream. Such liver delivery has been proposed to be particularly useful, for example, for expression of proteins that are missing in certain metabolic or hematological disorders, or that are effective in provoking immune responses (e.g., particularly antibody responses), for example against infectious agents or cancer cells. [0160] In some embodiments, administered LNPs are delivered to and/or taken up by antigen- presenting cells (e.g., as may be present in skin, muscle, mucosal tissues, etc.); such administration may be particularly useful or effective for induction of T cell immunity (e.g., for treatment of infectious diseases and/or cancers). [0161] In various embodiments, lipid nanoparticles can have an average size (e.g., mean diameter) of about 30 nm to about 150 nm, about 40 nm to about 150 nm, about 50 nm to about 150 nm, about 50 nm to about 130 nm, about 50 nm to about 110 nm, about 50 nm to about 100 nm, about 50 to about 90 nm, or about 60 nm to about 80 nm, or about 60 nm to about 70 nm. In some embodiments, lipid nanoparticles that may be useful in accordance with the present disclosure can have an average size (e.g., mean diameter) of about 50 nm to about 100 nm. In some embodiments, lipid nanoparticles may have an average size (e.g., mean diameter) of less than 80 nm, less than 75 nm, less than 70 nm, less than 65 nm, less than 60 nm, less than 55 nm, less than 50 nm, or less than 45 nm. In some embodiments, lipid nanoparticles that may be useful in accordance with the present disclosure can have an average size (e.g., mean diameter) of about 45 nm, 50 nm, 55 nm, 60 nm, 65 nm, 70 nm, 75 nm, 80 nm, 85 nm, 90 nm, 95 nm, 100 nm, 105 nm, 110 nm, 115 nm, 120 nm, 125 nm, 130 nm, 135 nm, 140 nm, 145 nm, or 150 nm. [0162] In some embodiments, lipids that form lipid nanoparticles described herein comprise: a polymer-conjugated lipid; a cationic lipid; and a helper neutral lipid. In some such embodiments, total polymer- conjugated lipid may be present in about 0.5-5 mol%, about 0.7-3.5 mol%, about 1-2.5 mol%, about 1.5-2 mol%, or about 1.5-1.8 mol% of the total lipids. In some embodiments, total polymer-conjugated lipid may be present in about 1-2.5 mol% of the total lipids. In some embodiments, the molar ratio of total cationic lipid to total polymer-conjugated lipid (e.g., PEG-conjugated lipid) may be about 100:1 to about 20:1, or about 50:1 to about 20:1, or about 40:1 to about 20:1, or about 35:1 to about 25:1. In some embodiments, the molar ratio of total cationic lipid to total polymer-conjugated lipid may be about 35:1 to about 25:1. [0163] In some embodiments involving a polymer-conjugated lipid, a cationic lipid, and a helper neutral lipid in lipid nanoparticles described herein, total cationic lipid is present in about 35-65 mol%, about 40-60 mol%, about 41-49 mol%, about 41-48 mol%, about 42-48 mol%, about 43-48 mol%, about 44-48 mol%, about 45-48 mol%, or about 46-49 mol% of the total lipids. In certain embodiments, total cationic lipid is present in about 47.0, 47.1, 47.2, 47.3, 47.4, 47.5, 47.6, 47.7, 47.8, 47.9 or 48.0 mol% of the total lipids. 11890709v1 24
Attorney Docket No.: 2013237-0929 [0164] In some embodiments involving a polymer-conjugated lipid, a cationic lipid, and a helper neutral lipid in lipid nanoparticles described herein, total neutral lipid is present in about 35-65 mol%, about 40- 60 mol%, about 45-55 mol%, or about 47-52 mol% of the total lipids. In some embodiments, total neutral lipid is present in 35-65 mol% of the total lipids. In some embodiments, total non-steroid neutral lipid (e.g., DPSC) is present in about 5-15 mol%, about 7-13 mol%, or 9-11 mol% of the total lipids. In some embodiments, total non-steroid neutral lipid is present in about 9.5, 10 or 10.5 mol% of the total lipids. In some embodiments, the molar ratio of the total cationic lipid to the non-steroid neutral lipid ranges from about 4.1: 1.0 to about 4.9: 1.0, from about 4.5: 1.0 to about 4.8: 1.0, or from about 4.7: 1.0 to 4.8: 1.0. In some embodiments, total steroid neutral lipid (e.g., cholesterol) is present in about 35- 50 mol%, about 39-49 mol%, about 39-46 mol%, about 39- 44 mol%, or about 39-42 mol% of the total lipids. In certain embodiments, total steroid neutral lipid (e.g., cholesterol) is present in about 39, 40, 41, 42, 43, 44, 45, or 46 mol% of the total lipids. In certain embodiments, the molar ratio of total cationic lipid to total steroid neutral lipid is about 1.5:1 to 1: 1.2, or about 1.2: 1 to 1: 1.2. [0165] In some embodiments, a lipid composition comprising a cationic lipid, a polymer-conjugated lipid, and a neutral lipid can have individual lipids present in certain molar percents of the total lipids, or in certain molar ratios (relative to each other) as described in WO 2018/081480, the entire contents of each of which are incorporated herein by reference for the purposes described herein. [0166] In some embodiments, lipids that form the lipid nanoparticles comprise: a polymer- conjugated lipid (e.g., PEG-conjugated lipid); a cationic lipid; and a neutral lipid, wherein the polymer- conjugated lipid is present in about 1-2.5 mol% of the total lipids; the cationic lipid is present in 35-65 mol% of the total lipids; and the neutral lipid is present in 35-65 mol% of the total lipids. In some embodiments, lipids that form the lipid nanoparticles comprise: a polymer-conjugated lipid (e.g., PEG-conjugated lipid); a cationic lipid; and a neutral lipid, wherein the polymer-conjugated lipid is present in about 1-2 mol% of the total lipids; the cationic lipid is present in 45-48.5 mol% of the total lipids; and the neutral lipid is present in 45-55 mol% of the total lipids. In some embodiments, lipids that form the lipid nanoparticles comprise: a polymer-conjugated lipid (e.g., PEG-conjugated lipid); a cationic lipid; and a neutral lipid comprising a non-steroid neutral lipid and a steroid neutral lipid, wherein the polymer-conjugated lipid is present in about 1-2 mol% of the total lipids; the cationic lipid is present in 45-48.5 mol% of the total lipids; the non-steroid neutral lipid is present in 9-11 mol% of the total lipids; and the steroid neutral lipid is present in about 36-44 mol% of the total lipids. In many of such embodiments, a PEG-conjugated lipid is or comprises a structure as described in WO 2017/075531 (also described
, or a some a PEG-conjugated lipid is or comprises 2-[(polyethylene glycol)-2000]-N,N-ditetradecylacetamide. In many of such embodiments, a cationic lipid is or comprises a chemical structure selected from I-1 to I-10 of Table 1 herein or a derivative thereof. In some embodiments, a cationic lipid is or comprises ((4-hydroxybutyl)azanediyl)bis(hexane-6,1-diyl)bis(2-hexyldecanoate). In many of 11890709v1 25
Attorney Docket No.: 2013237-0929 such embodiments, a neutral lipid comprises DSPC and cholesterol, wherein DSPC is a non-steroid neutral lipid and cholesterol is a steroid neutral lipid. [0167] In some embodiments, lipid nanoparticles include one or more cationic lipids (e.g., ones described herein). In some embodiments, cationic lipid nanoparticles may comprise at least one cationic lipid, at least one polymer-conjugated lipid, and at least one helper lipid (e.g., at least one neutral lipid). [0168] Figure 1 depicts a schematic of an exemplary drug manufacturing process 10. The process 10 may include the production of drug substance 12 followed by the production of drug product 14. Production of drug substance 12 may include in vitro transcription 16 followed by tangential flow filtration 18, followed by 0.2 micron filtration 20, followed by storage 22 at one or more temperatures in a range from about -20 deg C to about 8 deg C. Storage 22 may occur in a different location than the production of drug substance 12. After storage 22, drug subtance 12 may need to be transported to another location for production of drug product 14, which may include lipid nanoparticle (LNP) formation 24, followed by a second tangential flow filtration 26 process, followed by formulaion 28 and 0.2 micron filtation, folllowed by a second storage 30 process at one or more temperatures in a range from about -20 deg C to about 8 deg C. Following the second storage 30, the formulation may be transported to a different location for fill and finish steps including filling 32, visual insepction 34, labelling 36, and packaging 38. [0169] Figures 11-13 illustrate an exemplary LNP manufacturing process (for example, an RNA-LNP manufacturing process). [0170] Figure 11 illustrates an overview of exemplary manufacturing process 520 for a pharmaceutical-grade composition comprising RNA, according to aspects of the present disclosure. The process 520 may include the DNA transcription module 128, the first purification module 130, and the first bioburden reduction (or filtration) module 132, as previously described herein. In the embodiment of Fig. 11, the process 520 includes an exemplary manufacturing process for pharmaceutical-grade RNA comprising an in vitro RNA transcription followed by removal of components utilized or formed in the course of production by a purification process, and filtration to reduce bioburden (e.g., as illustrated in Figure 11). Optional in-process controls may also be completed depending on whether a hold step is performed. [0171] Figure 12 illustrates an overview of exemplary DNA template manufacture process 530 via a PCR-based process, according to aspects of the present disclosure. In the embodiment of Fig. 12, the process 530 includes an exemplary manufacturing process of a DNA template via a PCR-based process including the DNA transcription module 128, the first purification module 130, and the first bioburden reduction (or filtration) module 132, as described herein. Initially, a master mix preparation is made. Subsequently, forward primer and vector are added. The PCR-mix is transferred into a reagent reservoir and a PCR plate was filled. A PCR is completed comprising an initial denaturation, a denaturation step, an annealing step, a final extension step for 20-30 (e.g., 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) cycles and a hold step. The PCR products can be pooled and purified. Subsequently, the purified, pooled PCR product may be filtered and quality control tested. Fig. 12 illustrates which portions and/or steps of the process 530 are contained within each module (i.e., 128, 130, 132) of the process flow shown in Fig. 11. [0172] Figure 13 illustrates an exemplary process 800 for manufacturing LNP compositions. Generally, steps 806, 808, and 810 (as well as equipment associated with those steps, as described herein) will 11890709v1 26
Attorney Docket No.: 2013237-0929 occur and/or be located in the LNP formation module 70, shown in Fig. 5. Generally, steps 812 and 814 (as well as equipment associated with those steps, as described herein) will occur and/or be located in the second purification module (i.e., the second tangential flow filtration module) 80, shown in Fig.8. Generally, step 816 (as well as equipment associated with step 816, as described herein) will occur and/or be located in the second bioburden reduction module (i.e., formulation) 90 shown in Fig. 9. Steps following 816 may occur in other modules and/or may occur at other facilities (or not at all). For example, as explained herein, freezing and warehousing may not be required in all embodiments. [0173] Referring still to Fig.13, as can be seen, the produced compositions are prepared by combining lipids 810 with an aqueous preparation which carries an agent of interest (e.g., an active agent). In many embodiments, the agent of interest is a nucleic acid (e.g., a nucleic acid therapeutic). As depicted in Fig. 13, the nucleic acid is an RNA (e.g., a therapeutic RNA); in many embodiments of this depicted process, a utilized RNA includes at least one open reading frame (ORF) which may, for example, encode a vaccine antigen, a replacement protein, an antibody agent, a cytokine, etc.). In some embodiments a vaccine antigen may be a cancer vaccine antigen or an infectious disease (e.g., viral) antigen. In some embodiments, an RNA encodes a polypeptide that is or comprises a viral antigen such as a coronaviral antigen, such as a spike protein or portion thereof, or relevant variant of the foregoing (e.g., a SARS-CoV-2 spike protein or receptor binding domain thereof, for example, a prefusion stabilized variant thereof), e.g., as is utilized in one or more of mRNA- BNT162a1, mRNA-BNT162b1, mRNA-BNT162b2, mRNA-BNT-162c1, mRNA-1273, CVnCov, CVnCoV2, etc.). In certain embodiments exemplified herein, utilized was an RNA of BNT162b2. [0174] In some embodiments of the process depicted in Fig. 13, the RNA is prepared by in vitro transcription (e.g., of a DNA template which may, for example be a linear template such as a linearized plasmid or an amplicon). [0175] Referring to Fig. 13 and the exemplary process that it depicts, at step 808, the process 800 may include LNP formation by adding lipids 810 to an RNA solution 806, as well as high impact mixing (for example, via impingement jet mixing), and stabilization. Typically, the RNA solution is an aqueous solution. [0176] In many embodiments, the lipids 810 may include one or more of a cationically ionizable (sometimes referred to as “cationic” for simplicity) lipid, a phospholipid, a PEG-lipid, a sterol (e.g., a cholesterol) and an appropriate solvent (e.g., ethanol). [0177] In some embodiments, LNP formation may be performed in presence of a buffer (e.g., a citrate buffer) 812. In some embodiments, the buffer (e.g., a citrate buffer) 812 may be present in the RNA solution 806 prior to mixing with the lipids 810 (for example, via in-line dilution of the water-diluted RNA with the buffer (e.g., citrate buffer) 812 to form the aqueous solution of RNA 806). Stated otherwise, buffer (e.g., citrate buffer) 812 may be added to the RNA solution prior to mixing with the lipid solution 810. In some embodiments, the buffer (e.g., citrate buffer) 812 may also (or alternatively) be added to the mixture resulting from combining the lipid solution with the aqueous solution 806 (which, as depicted in Fig. 13, is an RNA solution but could, in some embodiments, carry a different agent). In some embodiments, the buffer (e.g., citrate buffer) 812 may include citric acid (monohydrate sodium citrate) and/or sodium hydroxide. [0178] According to embodiments described herein, step 808 (LNP formation) includes reducing or eliminating the introduction of various impurities into the process and/or various solutions thereof, thereby 11890709v1 27
Attorney Docket No.: 2013237-0929 forming a first RNA-LNP preparation that includes LNP-encapsulated RNA. LNP formation 808 may include the adjusting of one or more process temperatures, process flow rates, and/or ratios of the buffers, solutions and/or suspensions. LNP formation may include independently flowing each of the aqueous solution and lipids 810 (for example, in a lipid solution) into a mixing unit. Each of the aqueous RNA solution 806 and lipid solution 810 may flow into the mixing unit under laminar flow conditions (to avoid the entrapment of gas bubbles (for example, ambient gas bubbles (i.e., primarily comprised of nitrogen)) within the flow). [0179] Still referring to Fig.13, at step 814, the process 800 may include buffer exchange and concentration of the first RNA-LNP preparation to form a second RNA-LNP preparation. The buffer exchange and concentration step 814 may be conducted with process parameters including, for example, a feed flow rate, for example within a range of 18 to 50 liter/min (LPM), a trans-membrane pressure (TMP), for example lower than 1200 mbar, a retentate pressure, for example within a range of 130 to 230 mbar, and a permeate pressure, for example within a range of 10 to 70 mbar. [0180] In some embodiments, buffer exchange 814 of the first RNA-LNP preparation and concentrating the first RNA-LNP preparation are performed in alternating steps. In one or more embodiments, a TRIS (i.e., tris(hydroxymethyl)aminomethane) buffer may be used. In some embodiments, the buffer exchange 814 is conducted via diafiltration and the concentration is conducted via ultrafiltration. In some embodiments, the diafiltration and/or the ultrafiltration are conducted via tangential flow filtration (TFF) (for example, in a tangential flow filtration unit and/or TFF skid). In some embodiments, the tangential flow filtration is conducted using one or more jejunostomy tubes and/or one or more dip tubes configured to avoid introducing ambient gas bubbles into the second RNA-LNP preparation. During the tangential flow filtration, a retentate may be recirculated to a feed tank using a dip tube comprising a first end submerged into filtration feed liquid in the feed tank to avoid introducing ambient gas bubbles into the filtration feed liquid. Prior to the buffer exchange and concentration steps, a filtration system for tangential flow filtration may be filled with a buffer to prevent introducing ambient gas bubbles into the second RNA-LNP preparation. [0181] Referring still to Fig.13, the buffer exchange and concentration step 814 may include at least two buffer exchanges conducted via diafiltration alternating with at least two concentrations conducted via ultrafiltration. During buffer exchange and concentration 814, process temperatures may be maintained within a desired temperature range (for example, at or below about 25 deg C, or from about 2 deg C to about 25 deg C, or from about 15 deg C to about 25 deg C). During buffer exchange and concentration 814, pH may be continuously monitored (and may be maintained in a target range (for example, from about 7.0 to about 7.5, or from about 7.1 to about 7.3)) and shear may be maintained, for example in a range from about 2000 s^-1 to about 6000 s^-1, or from about 3000 s^-1 to about 5000 s^-1, or at about 4000 s^-1 (+/- 1%, 5%, and/or 10%). Following buffer exchange and concentration 814, a recovery flush may be performed, during which time shear may be reduced to under about 2000 s^-1 (for example, under about 1500 s^-1, or under about 1000 s^-1). In some embodiments, following buffer exchange 814, the pH may be maintained within a range from about 7.3 to about 7.5, for example following ultrafiltration and/or diafiltration. [0182] In some embodiments, during buffer exchange and/or concentration 814, the pH of the first RNA-LNP preparation may be maintained at a pH that is higher than that of the cationic lipid (i.e., the cationic lipid in the lipid solution). Without wishing to be bound by any particular theory, it is proposed that doing so may reduce foaming of the liquid nanoparticles. 11890709v1 28
Attorney Docket No.: 2013237-0929 [0183] In some embodiments, the first and/or second RNA-LNP preparation(s) may be sterilized without introducing any ambient gas into the produced formulation. In some embodiments, a relevant produced formulation may be a product for further manipulation, processing, packaging, and/or shipping. In some embodiments, a produced formulation may be or comprise a drug product formulation, e.g., for administration to humans. [0184] In some embodiments, one or more sterilization steps may be performed by sterile filtration; in some embodiments, sterile (or other) filtration may be conducted at a target pressure with substantially no pressure building up during the filtration process, for example at about 1.03 bar (or from about 1.02 bar to about 1.04 bar, from about 1.01 bar to about 1.05 bar, or from about 1.00 bar to about 1.1 bar). [0185] In some embodiments, a utilized mixing unit may include one or more impingement jet mixing skids. Prior to mixing, the impingement jet mixing skids may be vented and/or flooded to remove gas bubbles from tubing of the impingement jet mixing skids. Mixing of the aqueous and lipid solutions may be performed within boundaries of the mixing unit and/or one or more impingement jet mixing skids. In some embodiments, prior to mixing, the aqueous solution does not contact the lipid solution. In some embodiments, the flow rate ratio into the mixing unit of the aqueous solution to the lipid solution is about 3:1, or from about 2.75:1 to about 3.25:1, or from about 2.5:1 to about 3.5:1, and/or from about 2.5: to about 3:1. In some embodiments, the mixing speed may be adapted to avoid entrapping ambient gas in the first RNA-LNP preparation. For example, in order to avoid the introduction of ambient gas (and/or other impurities), one or more mixing processes may include increasing the mixing speed gradually until a slight vortex has formed (for example, the mixing speed at or slightly above the point at which a visible vortex has formed), but below the mixing speed at which foam begins to form. [0186] Still referring to Fig.13, the system (for example, the impingement jet mixing skids, the TFF system (i.e., the tangential flow filtration unit), and/or components thereof) may be assessed at one or more time points (e.g., monitored over time, e.g., periodically or continuously) for presence of gas bubbles. In the event that gas is detected in the aqueous solution, the lipid solution, the first RNA-LNP preparation, the second RNA-LNP preparation, the mixing unit, and/or tubing providing the aqueous solution of RNA, the lipid solution, the first RNA-LNP preparation, and/or the second RNA-LNP preparation, an alert or notification may be sent indicating that gas has been detected somewhere in the system. In some embodiments, gas detection may be performed via one or more flowmeters (for example, via one or more Coriolis flowmeters), and/or by visual assessment (e.g., via the human eye and/or various types of cameras), viand/or other detection means. [0187] In some embodiments, the aqueous solution and/or the lipid solution may be flowed into the mixing unit through one or more inlets disposed at a bottom portion of the mixing unit, and the resulting first RNA-LNP preparation may be released from the mixing unit through one or more outlets disposed at a top portion of the mixing unit. In some embodiments, the mixing may be performed with a submerged mixer. In some embodiments, foam may be generated during and/or after formation of the LNP-encapsulated RNA, and may be subsequently removed from the RNA-LNP preparation (for example, the foam may be removed from the first and/or second RNA-LNP preparation). [0188] Referring still to Fig.13, following buffer exchange 814 and concentration, the process 800 may include 0.2 µm filtration and/or the addition of sucrose and PBS for compounding. Following compounding, the process 800 may include bioburden reduction filtration (BBR) 816 following the buffer 11890709v1 29
Attorney Docket No.: 2013237-0929 exchange and concentration 814. Bioburden reduction filtration 816 may include filtering with 0.2 µm pore size (or for example, about a 0.22 µm pore size) or smaller filter. Bioburden reduction filtration 816 may also include using other pore sizes (for example, 0.45 µm pore size) as described herein. According to the present embodiments, 0.2 µm pore size filtering may also occur on each of the lipid solution and the aqueous RNA solution prior to mixing, on the first RNA-LNP preparation, and/or on the second RNA-LNP preparation. Bioburden reduction filtration 816 may also include filtering the post TFF-LNP suspension through a particulate reduction filter prior to filtering the suspension through (for example) the 0.2 µm pore size and/or 0.22 µm pore size bioburden reduction filter. In some embodiments, bioburden reduction filtration 816 may also include performing a filter recovery flush. [0189] Still referring to Fig.13, following bioburden reduction filtration 816, the process 800 may include filling transport bags (for example, Flexsafe ® bags) with the filtered second RNA-LNP preparation, and performing a visual inspection 818 of the transport bags for gas bubbles. In some embodiments, transport bags may be, for example 12 L bags, 50 L bags, 100 L bags, and/or other suitable bag sizes (e.g., depending on the batch size of the relevant RNA-LNP preparation), including bags that include a volume between 12 L and 50 L, and/or bags that include a volume between 50 L and 100 L. [0190] In some embodiments, filling transport bags may include filling the bags to a volume in a range from about 30% to about 95%, or from about 40% to about 90%, or from about 50% to about 85%, or from about 60% to about 85% or from about 70% to about 85%, and/or other subranges therebetween of the total bag volume. [0191] Filled bags may be stored and/or shipped at a temperature in a range from about 1 deg C to about 15 deg C (for example, at about 2 deg C to about 10 deg C, or from about 2 deg C to about 8 deg C), or alternatively may be frozen to a temperature of about -70 deg C (for example, in a range from about -60 deg C to about -80 deg C). Prior to shipment, the bags may be secured in or on racks and/or within or on any other suitable shelving or storage system so as to minimize movement, rupturing, and/or disruption of the bags during the transport to a fill and finish site. For example, transport bags may be stacked in a specific manner using a stacking system on pallets that include shock absorbers. During transport 820 and/or in preparation for transport 820, as well as following transport, nitrogen with a positive pressure (for example, from about 1-2 bars) may be maintained in and around the environment in which the bags are kept and/or transported, in order to prevent ambient gas from entering the bags. After transport 820, the bags may be assessed for ambient gas content (e.g., visually inspected) 822 a second time. In some embodiments, ambient gas bubbles that are discovered during such second assessment 822 may be removed (e.g., may be manually removed), or alternatively, the bag or bags that include ambient gas bubbles may be selectively discarded (for example, if the volume of ambient gas within a given bag has exceeded a threshold). [0192] Referring still to Fig.13, after arriving at a fill and finish site, sterile filtration 824 may be performed (i.e., the second RNA-LNP preparation). In some embodiments, such sterile filtration 824 may be performed after the preparation has been removed from the transport bags, but prior to being disposed within a collection vessel, reservoir, and/or fill tank. In some embodiments, the material (i.e., the filtered preparation) may then be dispersed from the collection vessel, reservoir, and/or fill tank during aseptic fill and finish 826 (for example, to aseptically fill glass vessels with the drug product). 11890709v1 30
Attorney Docket No.: 2013237-0929 [0193] Visual inspection 828 may be performed on the filled glass vessels. The inspected and filled glass vessels, at step 830 of the process 800, may then be frozen, stored, warehoused and/or distributed, for example, to health care administration sites. Alternatively, in some embodiments, filled glass vessels may be subjected to lyophilization or other drying process, so that drug product is transported and/or stored in a dry state (e.g., for subsequent resuspension). [0194] Still referring to Fig. 13, in some embodiments, the fill and finish facility may be located in the same location as the LNP production facility, in which case fill and finish may be performed directly using Point of Fill filtration equipment (in which case the transport 820, bag filling and sealing, and one or more of the visual inspection steps 818, 822, 828 may not be required. In yet other embodiments, the process 800 may include multiple transport steps 820, as well as additional visual inspection steps 818, 822, 828 if the various steps of the process 800 are performed at additional and/or other facilities (or alternatively, if transport is required within a single facility). [0195] Drug processing and preparation of lipid ethanol solutions, lipid nanoparticle (LNP) preparations (including RNA-LNPs), and other bioprocessing or chemical processing methodologies often require heating and cooling. Accordingly, the jacket of the present embodiments may be used as part of in vitro transcription (i.e., in connection with the bioreactors), tangential fill filtration, formulation, impingement jet mixing of lipids and RNA solutions, as well as during other processes. [0196] According to the present embodiments, the jackets may be cooled, heated, and/or pressurized. The internal fluid flow may also be adjusted. In some embodiments, the bottles and jacket rest on a table that has a magnetic stirrer (i.e., a “stirring plate” or “stirrer plate”). A bottom indentation in the bottle allows for impeller (or stirrer) to be seated on a hub around which it can rotate as a result of a magnetic coupling with a stirring plate in the table. In some embodiments, the single use and/or multi-use bottles may be made from polypropylene, as well as other suitable materials. In some embodiments, the jacket may be formed from stainless steel and/or other metallic or other materials that are sufficiently conductive to heat. The jacket may include a temperature control unit to maintain the internal temperature at the desired levels. In some embodiments, the jacket may include one or more clamps located at the fluid inlets and outlets, which may be turned to enable thermal fluid flow into and out of the vessel. The jacket may include a fluid inlet and a fluid outlet, for routing cooling and/or heating fluids in and out. [0197] In some embodiments, cooling and/or heating fluids that are used in connection with the jacket may include water, silicon oil, ethanol, and/or other thermal fluids. In some embodiments, the jacket may include one or more sensors (for example, temperature probes such as RTDs, thermocouples, other types of temperature probes, pressure sensors, differential pressure sensors, flow sensors, etc.). In some embodiments, flow enters at the bottom and leaves at the top, along with the air. In some embodiments, the jacket includes two sides, shells, and/or halves such that the jacket may be opened and closed. In some embodiments, one side, shell, or half of the jacket may be fixed to a table. The two shells may be connected via hinges such that one side, half, or shell rotates about the one or more hinges relative to the second side, half, or shell. The two sides, halves, and/or shells may be hinged into a closed position and attached together via one or more attachment means such as a latch, buckles, clasp, bayonet feature, and/or other suitable attachment means. In some embodiments, the attachment means is non-magnetic so as to prevent any interference with the heating / cooling fluid and/or the contents within the bioprocessing vessel / bioreactor 11890709v1 31
Attorney Docket No.: 2013237-0929 and/or single-use bottles. In some embodiments, one or more programmable limit switch (PLS) controllers may be used to control the magnetic stirred speed and check the temperature. In some embodiments, the bottle may be composed of polypropylene and may be configured to accommodate internal pressures up to about 1 bar and/or in some embodiments, up to 5-10 bars. In some embodiments, the bottle may have an internal volume of about 1 L to about 50 L, or from about 2 L to about 5 L or from about 10 L to about 20 L, or from about 20 L to about 50 L, or from about 2 L to about 20 L, or from about 4 L to about 50 L, or from about 5 L to about 20 L, or from about 5 L to about 10 L, and/or other subranges therebetween. [0198] In some embodiments, the jacket is used to heat to temperatures in the vicinity of 35 deg C, or from about 30-40 deg C, while also being configured to cool to temperatures in a range from about 2 deg C to about 8 deg C. In some embodiments, the jacket may be used to provide a wide temperature range, for example, temperatures in a range from 0 deg to 100 deg, or from about 1 deg C to about 99 deg C. In some embodiments, the jacket is shaped (i.e., includes internal contouring and/or features) to match and/or accommodate the size, shape, and/or features of a particular single use bottle, bioreactor, and/or other bioprocessing vessel. In some embodiments, the jacket may be used in connection with bioreactors and/or single use bottles that include one or more inlets at the top (for example, for adding lipids or other materials, as well as one or more additional openings for siphoning off portions of the mixture contained therein. In some embodiments, the jacket may be used in connection with a bottle or vessel (for example, a bioreactor) that includes both a bottom stirrer and a top stirrer (for example, magnetically actuated stirrers disposed in the interior of the bottle or bioreactor with one or more corresponding electromagnetic inducers disposed within the bottom surface and/or top surface of the bottle and/or bioreactor, such that the electromagnetic inducers are magnetically coupled to the respective magnetic stirrer). [0199] In some embodiments, the jacket may be used in connection with one or more drug production processes, for example in the preparation of a lipid-ethanol solution. In some embodiments, the jacket may be used in connection with a tangential flow filtration process and/or in connection with a drug formulation process. In some embodiments, the jacket can be used in any application where heating, cooling, and/or stirring are required. In some embodiments, the jacket may be used in connection with a buffer preparation process. In most embodiments, the jacket does not need to be cleaned after each use because the jacket includes a closed loop system for circulating one or more cooling or heating fluids, as described herein. The closed loop system(s) within the jacket are fluidly disconnected from the interior of the single-use bottle, bioreactor and/or bio-processing vessel. Accordingly, there is low to no risk of cooling/heating fluid from the closed-loop systems within the jacket from impurifying and/or otherwise contaminating the processes within the interior of the single use bottle / bioreactor. Each of the jackets of the present embodiments may be used to accommodate a number of processes with varying process parameters. For example, in a first use, the jacket could be used to heat a bioreactor being used in an in vitro transcription process. In the very next use, the same jacket could be used to cool a single use bottle or vessel being used to mix ethanol and lipids. In both cases, the jacket provides the desired conditions (i.e., temperatures) to the vessel positioned within in it via the temperature control unit (which also dictates the fluid pressure and flow rate). The inlet and/or outlet may include one or more sealing features (for example, rubber gaskets, O-rings, quick-connects, and/or other features) at the interface between the internal channels of the jacket and the fluid lines connected thereto. For example, in some embodiments, the jacket may include a pair of semi-circulate plates or outer casings disposed around the outer periphery of the jacket (for example, one semi-circulate plate or casing disposed around each 11890709v1 32
Attorney Docket No.: 2013237-0929 of the two jacket shells) to seal the channels (i.e., forming the radially outer walls of the channels). The semi- circulate plates or outer casings may be composed of insulating materials (i.e., rather than heat-conducting materials) so as to discourage heat transfer between the jacket / channels and the external environment. In some embodiments, where heat transfer to the environment is less of a consideration (i.e., in connection with smaller jackets) the outer casing may be composed of a metallic material such as stainless steel. In some embodiments, where heat transfer to the environment is more of a consideration (i.e., in connection with larger jackets) the outer casing may be composed of an insulting material such as a structural polymer or foam. According to aspects of the present embodiments, an important consideration includes selecting structural polymers or other insulating materials that are not reactive with the thermal fluids. As such, in some embodiments, the outer casing includes a composite structure that includes a metallic (i.e., stainless steel) inner layer and an insulating (i.e., polymer or foam) outer layer. [0200] In some embodiments, the jacket may include one or more grooves to allow for tubing (to allow inlet / outlet flows) to be disposed therethrough, the tubing connecting an interior of the vessel, bottle, or bioreactor to an exterior of the vessel, bottle, or bioreactor. In some embodiments, internal features of the jacket may be made via casting, forging, 3D printing / additive manufacturing, machining, fabrication, milling, CNC drilling, electrical discharge machining (EDM), and/or other suitable processes. In some embodiments, the jacket may include an outer diameter of about 28 cm to accommodate a 10 L bottle / bioreactor. In some embodiments, the jacket may include other dimensions and/or may accommodate a different size vessel or container. In some embodiments, the jacket may be produced by starting with a stainless-steel cube and using one or more milling and / or computer numerical control (CNC) processes (for example, drilling using a 5-axis machine) to form the exterior and interior shapes including channels, protrusions, grooves, bores, etc. In some embodiments, the outer diameter may be smaller or larger to accommodate smaller or larger bottle sizes. In some embodiments, the internal size and number of channels and/or channel passes may be scaled up and down in order to optimize the heating and/or cooling effectiveness. [0201] Figure 2 depicts an overview of an automated drug manufacturing process 40, according to aspects of the present disclosure. The jacket of the present invention may be used in connection with the drug manufacturing process (and various sub-processes and equipment used thereine) of Fig.2. The process may include in vitro transcription 50, a first tangential flow filtration process 60, LNP formation 70, a second tangential flow filtration 80, formulation 90, and fill and finish 100. The automated drug manufacturing process 40 of the present embodiments (illustrated in Fig. 2) may include a fully automated process occurring all at a single site (for example, at a single location within a single, contiguous footprint (i.e., production facility) comprising a total area of no more than about 8,000-12,000 square meters (for example, within an area comprising from about 100 square meters to about 12,000 square meters, or from about 200 square meters to about 10,000 square meters, or from about 300 square meters to about 8,000 square meters, or from about 200 square meters to about 6,000 square meters, or from about 500 square meters to about 3,000 square meters, or from about 1,000 square meters to about 5,000 square meters, or from about 1,500 square meters to about 4,000 square meters, or from about 750 square meters to about 3,500 square meters, or from about 500 square meters to about 1,750 square meters, and/or other subranges therebetween). As such, several steps of the exemplary process 10 illustrated in Fig. 1 (such as transport between locations, and multiple storage (and thawing) steps) may be able to be eliminated. In addition, because the process 40 is fully automated, less manpower is required to run the process. For example, the process may be monitored 11890709v1 33
Attorney Docket No.: 2013237-0929 remotely, with no human intervention required to run the process once it has been supplied with electricity, a water supply, and/or the necessary input materials. [0202] Figures 3A and 3B illustrate perspective views of single use bottles 52 in a heating and cooling jacket 42, according to aspects of the present disclosure. Each of the heating and cooling jackets 42 (i.e., “jackets”) may include first and second halves, portions or shells (i.e., “ shells”) 46, 48. Each of the first and second shell 46, 48 may be substantially semi-cylindrical such that the two shells 46, 48 together may form a jacket 42 (for example, a cylindrical jacket) around a single use bottle 52. According to aspects of the present embodiments, each jacket 42 may be used in connection with single use bottles 52, as well as other bioprocessing vessels such as bioreactors, mixing vessels, reservoirs, etc. As shown in Fig.3A, the first and second shells 46, 48 may come together along an interface 78 such that the jacket 42 tightly encapsulates the single use bottle 52. The jacket 42 may include a joining means 76 (for example, a clasp, latch, buckle, etc.) for securing the first and second shells 46, 48 together. As shown in Fig.3B, the first and second shells 46, 48 may hinge relative to one another about a hinge axis 68. The jacket 42 may include one or more hinges that are disposed co-linearly with the hinge axis 68, the one or more hinges not being visible in the illustration of Fig. 3B. [0203] Referring still to Fig. 3, each jacket 42 may be disposed on a support table 44 that may be used to support the jacket 42 and single use bottle 52. In some embodiments, the support table 44 may also be used to actuate (for example, rotate) a stirrer or impeller 89 (shown in Fig. 5A) positioned in the bottom of the single use bottle 52 (or bioprocessing vessel 52). The table 44 may include local controls 64 and/or a control unit for controlling the speed of the stirrer. In some embodiments, the table 44 includes a magnetic field generator and/or an inducer that is/are magnetically coupled to the stirrer 89, thereby enabling articulation of the stirrer (or impeller) 89 by the table controls 64. In some embodiments, the single use bottle 52 may include a second stirrer or impeller 89 disposed near a top surface 62 of the single use bottle, the second stirrer also being disposed within the interior of the single use bottle 52. In some embodiments, the second stirrer (near the top of the single use bottle 52) may be mechanically coupled (for example, via a shaft aligned longitudinally along a center axis of the single use bottle 52) to the first stirrer (at the bottom of the single use bottle 52) to further enhance mixing of the substances within the single use bottle 52. In some embodiment, the second stirrer is instead coupled to a second magnetic field generator or inducer (not shown) located adjacent to (or in the vicinity of) the top surface 62 of the single use bottle 52. In some embodiments, one half of the jacket 42 (for example, first shell 46 or second shell 48) may be firmly attached to the table while the other half of the jacket 42 may be free to rotate about the hinge axis 68, thereby allowing the jacket 42 to be opened and closed for insertion and removal of the single use bottle 52. Firmly attaching one half of the jacket 42 to the table 44 helps to prevent the likelihood of the bottle 52 and/or jacket 42 from falling off of the table 44. [0204] Still referring to Fig. 3, each jacket 42 may include one or more inlets 66 and one or more outlets 54 for routing thermal fluids (such as silicone oil, water, mixes thereof, etc.) into and out of the jacket 42 for cooling and/or heating the contents of the bioprocessing vessel 42 (or single use bottle 42). In some embodiments, the one or more inlets 66 are disposed at the bottom of the jacket 42 while the one or more outlets 54 are disposed at the top of the jacket 42, as shown in Fig. 3A. In some embodiments, the one or more inlets 66 are disposed at the top of the jacket 42 while the one or more outlets 54 are disposed at the bottom of the jacket 42, as shown in Fig. 3B. In some embodiments, placing the one or more inlets 66 at the 11890709v1 34
Attorney Docket No.: 2013237-0929 bottom and the one or more outlets 54 at the top of the jacket 42 helps to encourage any trapped gas and/or air bubbles make their way out of the jacket 42, thereby preventing the gas(es) from building up in the interior of the jacket 42. Each of the one or more inlets 66 and one or more outlets 54 may be fluidly coupled (via any suitble means) to external tubing (not shown) for routing flows into and out of the jacket 42. In addition, as disclosed herein, each of the inlets 66 and outlets 54 may be fluidly connected to one or more channels and/or passageways or passages disposed within the interior of the jacket 42. Each jacket 42 may include one or more through-bores (for example, a small circular through-bore 72, a larger, elongated through-bore 82 (i.e., with rounded corners in some embodiments), as well as other through-bores, in various shapes and sizes) such that nipples 74, adaptors, couplings and/or other connections disposed within the single use bottle 52 may be connected to tubing that is positioned external to the jacket 42. Each single use bottle 52, may include one or more openings disposed in the top of each bottle (for example, a smaller opening 56 and a larger opening 58 for adding materials to and/or siphoning or suctioning materials from the the single use bottle 52). The small opening 56 and large opening 58 may be in fluid communication with nipples 74, adaptors, couplings and/or other connections disposed in the single use bottle 52 such that a separate flow circuit is formed (i.e., a second or third flow circuit) that is different from, and fluidly disconnected from the one or more cooling / heating flow circuits disposed within the jacket 52 as described herein. [0205] Figure 4 illustrates a support table 44, according to aspects of the present disclosure. The table 44 may include a platform 84 for supporting the single use bottle 52 (and/or other bioprocessing vessel 52) as well as the local controllers 64 (i.e., for adjusting the speed at which the stirrer or impeller 89 (shown in Fig. 5A) spins within the single use bottle 52). The platform 84 may also include a magnetic field generator or inducer (for example, located beneath the platform 84) for magnetically coupling to and actuating (i.e., rotating) the stirrer or impeller 89 within the single use bottle 52 (or bioprocessing vessel 52). [0206] Figures 5A and 5B illustrates details of a single use bottle 52 in a heating and cooling jacket 42, according to aspects of the present disclosure. Fig.5A illustrates a cross-section of a single use bottle 52 disposed within a jacket 42, while Fig.5B illustrates an interior perspective view of a jacket shell 48 (for example, a second jacket shell 48 with a small circular through-bore 72 and a larger elongated through-bore 82 disposed therethrough) according to aspects of the present embodiments. The jacket 42 may include a plurality of channels 86 that extend circumferentially around the jacket and interconnect with each other. For example, each of the two shells 46, 48 that form the jacket 42 include an interconnected network of channels 86 or passages that extend circumferentially around (and within) the shell 46, 48 and connect to a channel that is longitudinally adjacent to each (for example, each channel 86 connects to the channel above and/or below it in the view illustrated in Fig.5A). The connections 124 between channels 86 are shown in Fig. 6. The plurality of channels 86 fluidly connect the fluid inlet 66 to the fluid outlet 54 within each of the shells 46, 48. As such, each of the first shell 46 and the second shell 48 includes a flow circuit from the fluid inlet 66, to the plurality of channels 86, to the fluid outlet 54. The fluid circuits (for example, a first fluid circuit and a second fluid circuit) within each of the first shell 46 and the second shell 48, respectively, do not join together within the jacket 42. [0207] Referring still to Fig. 5, the table 44 includes the magnetic field generator 88 (or inducer 88) disposed beneath the surface of the table, and in some embodiments may also include a control unit 92 (i.e., table control unit 92) for controlling the magnetic field generator 88 (or inducer 88), as illustrated in Fig. 5A. In some embodiments, the control unit 92 may wirelessly (or via a wired connection) interface with other systems (for example, with a temperature control unit (TCU)). In some embodiments, the table 44 may include a 11890709v1 35
Attorney Docket No.: 2013237-0929 plurality of supports 94 that extend vertically downward as well as laterally beneath the magnetic field generator 88 (or inducer 88) and control unit 92, so as to support those components. As illustrated in Fig.5B, each of the first and second shells 46, 48 may include and/or be connected to one or more hinges 118 (for example, first hinge 118A and second hinge 118B, both being aligned with and centered on the hinge axis 68). The hinges 118A, 118B are illustrated as being attached to second shell 48 in Fig. 5B for illustration purposes, but would also connect to first shell 46. In some embodiments, each hinge 118 of the plurality of hinges 118 may be located above the elongated through-bore 82. [0208] Still referring to Fig. 5, the plurality of channels 86 may include channels 86 that include different radial widths 122 and/or that are disposed at different radii from a bottle centerline 120. The jacket 42 may include varying widths 122 and varying radii from the centerline 120 at which the channels 86 are disposed in order to accommodate varying outer diameter profiles 110, 112, 114, 116 of the single use bottle 52. For example, the single use bottle 52 may include one or more protruding portions 112, 116 that protrude radially outwardly beyond a nominal outer diameter 98 of the single use bottle 52 (i.e., which is also approximately a nominal inner diameter of the jacket 42). The single use bottle 52 may also include one or more recessed or concave portions 110 (when viewed externally) that extend radially inwardly to a radius that is less than the nominal inner diameter 98 of the jacket 42. The single use bottle 52 may also include one or more portions 114 (i.e., portions of the exterior surface of the bottle 52) that are disposed at a nominal outer diameter 98 of the single use bottle 52. The bottle 52 may include protruding portions 112, 116 to add structural robustness. The bottle 52 may include concave or recessed portions 110 to facilitate the easy of carrying the bottle (for example, by a forklift, human, or robot). To accommodate the varying outer diameter of the single use bottle 52 (i.e., portions with diameters that extend outward or inward of the nominal bottle outer diameter 98), the jacket may include corresponding protrusions and/or recesses. For example, recesses 104 and 108 in the inner surface of the jacket 42 help to accommodate protrusions 112 and 116 of the bottle 52. Similarly, protrusion 102 (which extends radially inward of the nominal inner diameter 98 of the jacket) helps to accommodate recessed portion 110 of the bottle 52. Portion 106 on the interior surface of the jacket 52 corresponds to portion 114 of the bottle 52, which is positioned at the nominal diameter 98. By contouring the interior surface of the jacket 42 such that it matches an exterior surface of the bottle 52, not only does the jacket 42 fit more snuggly around the bottle 52 (thereby reducing any air pockets trapped therebetween) but it also brings the heating and/or cooling fluid (and channels through which it is flowing) closer to the outer surface of the object (i.e., the single use bottle 52) to be heated or cooled. In addition, contouring the jacket to match the bottle increases the surface aware across which heat transfer may occur. Accordingly, an outer diameter 96 of the jacket 42 may be substantially constant / uniform (for example, within standard manufacturing tolerances) while the inner diameter may intentionally vary (as described herein) around a nominal inner diameter 98 of the jacket 42. Similarly, the plurality of channels 86 may include a constant (or uniform) outer radius about the centerline 120, and varying inner radii about the centerline 120. [0209] Figures 6A and 6B illustrates details of a heating and cooling jacket, according to aspects of the present disclosure. Figures 6A and 6B show illustrations of a second shell 48 (perspective and side views, respectively). However, with the exception of the circular through-bore 72 and the elongated through-bore 82, the remaining features would be present in the first shell 46 as well. As illustrated in Figues 6A and 6B, each of the channels runs circumferentially around the jacket (for example, around first jacket shell 46 and/or second jacket shell 48) and connect with adjacent channels 86 at connection channels 124 disposed at the ends of 11890709v1 36
Attorney Docket No.: 2013237-0929 each channel 86. A first plurality of channels 86A may include inner diameters that are more radially inward than the inner diameters of a second plurality of channels 86B, to accommodate bottles with varying outer diameters, as discussed herein in connection with Figs. 5A and 5B. The first and second shells 46, 48 may include a plurality of channel walls 142 defining the boundaries of each channel 86. Each channel 86 may also include a radially inner wall 140 that includes a semi-circulate contouring. As such, each channel extends in a semi-circular path around the jacket 42 and also includes a semi-circulare cross-section. [0210] Referring still to Figs.6A and 6B, an outer casing (not shown) that interfaces with lip 126 forms the radially outer walls of each channel 86. The outer casing may be welded, brazed, adhered, glued, compression fitted, mechanically attached, screwed into, and/or otherwise attached to the lip 126 and channel walls 142. The lip 126 extends around the exterior (i.e., radially outer) surface of each of the first and second shells 46, 48, and forms an interfacing surface for the outer casing to atttach to. The outer casing may be composed of metallic materials as well as one or more structural polymers that also acts as a thermal insulator such as polyethelene, polyvinylchloride, polypropylene, polyamide, as well as other suitable materials. The first and/or second shell 46, 48 may include a plurality of channels 138 with a shorter channel length 134 than the remainder of the channels 86, in order to accommodate and/or make space for the circular through-bore 72 and the elongated through-bore 82. The first and/or second shell 46, 48 may also include a vertically aligned manifold portion 136 for interfacing with the fluid inlet 66 (shown in Figs.3 and 5). [0211] Figures 7A and 7B illustrate a side view of a single use bottle 52 in a heating and cooling jacket 42 (Fig.7A) and a perspective view of a jacket 42 (for example, a second shell 48, Fig. 7B), according to aspects of the present disclosure. As shown in Fig.7A, the heating and cooling jacket 42, in some embodiments, includes an outer diameter 51 that is larger than that of the single use bottle 52, and a height 47 that is smaller than that of the single use bottle 52. As shown in Fig. 7B, the heating and cooling jacket 42 may include a sensor access port 49, for allowing a temperature sensor or other type of probe to be operatively coupled with the interior of the single use bottle 52 (i.e., thereby enabling the sensor to sense at least one operating parameter from the interior of the single use) while also allowing a wire or communication line 186 to extent outside of the heating and cooling jacket 42. In some embodiments, the temperature sensor or other type of probe may be inserted into a protrusion 59 (shown in Fig. 7A). [0212] Figure 7C illustrates details of a heating and cooling jacket sensor access port 49, according to aspects of the present disclosure. The sensor access port 49 may include a first rounded portion 53, a second rounded portion 55, and an elongated portion 57. In some embodiments, the elongated portion 57 separates the first rounded portion 53 from the second rounded portion 55. In some embodiments, the first rounded portion 53 includes a larger radius than the second rounded portion 55. In some embodiments, the first rounded portion 53 extends about 270 degrees (for example, from about 240 degrees to abuot 300 degrees, or from about 260 degrees to about 280 degrees) while the second rounded portion extends about 180 degrees (for example, from about 170 degrees to about 190 degrees). In some embodiments, the first rounded portion 53 is centered about a location of a centerline of the sensor or probe when it is inserted into the protrusion 59 (which protrudes externally from the single use bottle 42). The elongated portion 57 and second rounded portion 55 enable the second shell 48 to be rotated about the single use bottle 52 when the jacket 42 is being closed such that the protrusion 59 is disposed through the sensor access port 49. For example, elongated portion 57 and second rounded portion 55 make space for the protrusion 59, thereby enabling closure of the jacket 42 via rotation about the single use bottle 42. For example, if the sensor access 11890709v1 37
Attorney Docket No.: 2013237-0929 port 49 included only the first rounded portion 53 (i.e., and not elongated portion 57 or second rounded portion 55), closure of the heating and cooling jacket 42 may only be possible by moving the second shell 48 radially inward around the single use bottle 52, rather than rotating it into place, since the protrusion would interfere with an interior surface of the second shell 48. [0213] Referring still to Fig.7C, the sensor access port 49 may be dimensioned around the diameter and protrusion distance (i.e., the distance the protrusion 49 extends from the exterior of the bottle / vessel 52) of the protrusion 59, for each of the various jacket 42 sizes. The sensor access port 49 shown in Fig. 7C, in some embodiments, corresponds to a 10 L vessel / bottle 52, and may include, for example, a radius of about 9 mm for the first rounded portion 53, a radius of about 5.5 mm for the second rounded portion 55, and a third rounded portion (marked with “R3,0” in Fig. 7C) at a transition between the first rounded portion 52 and the elongated portion 57, the third rounded portion including a radius of about 3.0 mm. [0214] Figures 8A and 8B illustrate details of a fabricated / manufactured heating and cooling jacket (for example, of a second shell 48), according to aspects of the present disclosure. [0215] Figures 9A and 9B illustrate perspective views of single use bottles 52 in a heating and cooling jacket 42, according to aspects of the present disclosure. [0216] Figure 10 illustrates a system 150 for heating and cooling single use bottles 52 including the jacket 42, table 44, magnetic field generator 88 (or inducer 88), table control unit 92, fluid inlet 66, and fluid outlet 54, according to aspects of the present disclosure. The system 150 may include a pump 144 for pressurizing one or more cooling and/or heating fluids, as well as a pump suction line 170 and a pump discharge line 146. The system may include one or more temperature handling units 148 disposed fluidly downstream of the pump discharge line 146. The temperature handling unit 148 includes the capability to both heat and cool fluids (for example, thermal fluids such as silicon oil, water, glycol, water-glycol mixes, etc.). In some embodiments, the density of the thermal fluid does not exceed 1 kg/dm^3. In some embodiments, the thermofluid includes a water-glycol mixture or a water-ethylene-glycol mixture. In some embodiments, where cooling and heating is limited to a range of from about 2 deg C to about 35 deg C, a water-glycol mixture is used as the thermal fluid. In some embodiments, the thermal fluid includes a pH in a range from 6.0 to 8.5. For example, the temperature handling unit may include a heat exchanger that is in thermal communication with the fluid line(s) as well as both a heat source and a cooling source. The heat source may include a heater that uses liquid fuel as a heat source. In some embodiments, the heat source may additionally or alternatively include an electric heater (for example, an induction heater or a resistance heater). The cooling source may include a refrigeration unit (i.e., a cooling or coolant circuit) and/or a Peltier device (which is electrically powered and can act as both a heating source and cooling source). The temperature handling unit 148 may include one or more expansion vessels or reservoirs for storing thermal fluids. In embodiments that include a coolant circuit, the temperature handling unit 148 may include an evaporator upstream of a compressor, upstream of a condenser that is in turn positioned upstream of the evaporator. The temperature handling unit may also include one or more motors (i.e., one or more stepper motors), one or more pumps, stepper valves, and/or additional heat exchangers (for example, integrated into the evaporator and/or condenser). The system 150 may include one or more inlet lines (for example, first inlet line 152 and second inlet line 154) at the exit of the temperature handling unit 148 for delivering cooling / heating fluid to the inlets 66 of the jacket 42. In some embodiments, the system 150 includes two separate lines 152, 154 connecting the temperature handling 11890709v1 38
Attorney Docket No.: 2013237-0929 unit 148 to the inlets 66 for each of the first and second shells 46, 48 (shown in Fig.3A). In some embodiments, the system 150 includes only a single line connecting the temperature handling unit 148 to the inlets 66, with a splitter or T-junction disposed upstream of the inlets 66. [0217] Still referring to Fig. 10, according to aspects of the present embodiments, temperature handling units (or temperature control units (TCUs)) 148 from different manufacturers (such as Huber (i.e., Huber Unistat 410)) may be used. In some embodiments, a Huber Unistat 410, 410(w), or equivalent may be used in connection with heating and cooling of smaller jackets (i.e., 1 L, 2 L, 5 L, 10 L vessels) while a Unistat 530(w), 705(w) or equivalent may be used for large jackets (10 L, 20 L, 50 L vessels). [0218] Referring still to Fig. 10, the system may include one or more fluid outlet lines (for example, fluid outlet lines 158 and 160) for connecting the fluid outlets 54 to a downstream reservoir 166, which may be fluidly connected to the pump suction line 170. The system may include one or more valves 168 disposed in the pump suction line 170, as well as in other locations throughout the system. In some embodiments, a single line (for example, in connection with a T positioned at or near the two fluid outlets 54) may be used to delivery fluid to the reservoir 166 instead of the first and second fluid outlet lines 158, 160. In the side view illustration of Fig.10, only a single fluid inlet 66 and a single fluid outlet 54 are shown. However, each of the first shell 46 and the second shell 48 will have its own fluid inlet 66 and fluid outlet 54, as shown in Fig. 3A, and as disclosed herein. The system 150 may include one or more robots (for example, a first robot 162 and a second robot 164) for moving various components of the system as needed including but not limited to, the single use bottle 52, the jacket 42, as well as other components. [0219] Still referring to Fig. 10, the system 150 may include one or more sensors 156 operatively and/or communicatively coupled to an interior of the single use bottle 52 for sensing one or more characteristics thereof such as pressure, temperature, oxygen content, pH, polydispersity, viscosity, particle size, molecular weight, and/or other parameters. The system 150 may include a temperature control unit 172 communicatively coupled to each of the electronic components of the system including the pump 144, the temperature handling unit 148, the table control unit 92, the sensor(s) 156, the first robot 162, the second robot 164, and the valve 168, in addition to other potential system components. In some embodiments, the temperature control unit 172 may act as more of an overall control system as it is interacting with, and potentially sending commands to, various components that do not function in a way that affects the temperature of the interior of the single use bottle 52, etc. For example, in some embodiments, the temperature control unit (TCU) 172 may be controlled via a PLS / programmable limit switch, (for example, a Siemens PCS7 or equivalent with Ethernet / TCP/IP communication capabilities, for example, capable of operating with one or more of the following telecontrol protocols: SINAUT ST7, DNP3, Modbus RTU, IEC 60870-5-101, and IEC 60870-5-104). In some embodiments, the pump 144 may be located fluidly downstream of the temperature handling unit 148. [0220] Figure 11 depicts an overview of an exemplary manufacturing process for a pharmaceutical- grade composition comprising RNA that may include the use of the jacket 42, according to aspects of the present disclosure, and as discussed above. [0221] Figure 12 illustrates an overview of exemplary DNA template manufacture process via a PCR-based process that may include the use of the jacket 42, according to aspects of the present disclosure, and as discussed above. 11890709v1 39
Attorney Docket No.: 2013237-0929 [0222] Figure 13 illustrates an exemplary process for manufacturing LNP compositions that may include the use of the jacket 42, according to aspects of the present disclosure, and as discussed above. [0223] Figure 14 illustrates a method or process 900 for heating or cooling a chemical or biological process, according to aspects of the present disclosure. At step 902, the method 900 may include providing a bioprocessing vessel 52 (for example, a single use bottle and/or bioreactor according to the present disclosure) with a biological (or chemical) substance. At step 904, the method 900 may include disposing the bioprocessing vessel 52 within the the first jacket shell 46. At step 906, the method 900 may include rotating the second jacket shell 48 closed to enclose the bioprocessing vessel 52 within the jacket 42. At step 908, the method 900 may include securing the second jacket shell 48 to the first jacket shell 46 via latch (or locking means 76). At step 910, the method 900 may include setting initial process conditions via the temperature control unit such that thermal fluid temperature, pressure, and/or flow rate are adjusted to deliver the desired heating and/or cooling to the bioprocessing vessel 42. At step 912, in connection with step 910, the method 900 may include determining the flow rate, pressure, and/or temperature at which the thermal fluid will be circulated within the jacket 42. At step 914, the method 900 may include measuring at least one parameter within the bioprocessing vessel 42. At step 916, the method 900 may include setting a secondary set of process conditions (for example, different conditions than those set in step 910). At step 918, the method 900 may include determining the required flow rate, pressure and/or temperature at which the thermal fluid will be circulated within the jacket 42 to provide the secondary process conditions (i.e., temperature) within the bioprocessing vessel 52. At step 920, the method 900 may include measuring at least one process parameter within the bioprocessing vessel 52. At step 922, the method 900 may include repeating steps 910 through 920 (for example, including adjusting the conditions to third, fourth, fifth, etc. sets of conditions) until the overall bioprocessing process is complete. [0224] Referring still to Fig. 14, the first set of process conditions (step 910) may include a temperature that is above room temperature while the second set of process conditions (916) may include a temperature that is below room temperature, and vice versa. Therefore, the system described herein is capable of both heating and cooling. For example, a temperature below room temperature may include a temperate in a range from about 2 deg C to about 8 deg C, or from about 8 deg C to about 12 deg C, or from about 10 deg C to about 15 deg C, or from about 12 deg C to about 18 deg C, or from about 2 deg C to about 18 deg C, or from about 2 deg C to about 17 deg C, or from about 1 deg C to about 17 deg C, and other subranges therebetween. A temperature above room temperature may include a temperate in a range from about 26 deg C to about 30 deg C, or from about 30 deg C to about 34 deg C, or from about 35 deg C to about 40 deg C, or from about 32 deg C to about 37 deg C, or from about 26 deg C to about 40 deg C. In some embodiments, the temperature set at steps 910 and 912 may be at least 15 deg C different than the temperature set at steps 916 and 918. In some embodiments, the temperature set at steps 910 and 912 may be at least 20 deg C different than the temperature set at steps 916 and 918. In some embodiments, the temperature set at steps 910 and 912 may be at least 25 deg C different than the temperature set at steps 916 and 918. In some embodiments, the temperature set at steps 910 and 912 may be at least 30 deg C different than the temperature set at steps 916 and 918. The system described herein may employ the same thermal fluid (water, silicon oil, mixes thereof, etc.), for both cooling and heating. According to aspects of the present embodiments, the steps illustrated in Fig. 14 may be performed in a different order than what is shown. In some embodiments, the method 900 may include additional steps (i.e., steps not shown in Fig. 14). In some 11890709v1 40
Attorney Docket No.: 2013237-0929 embodiments, not every step shown in method 900 is performed. In some embodiments, one or more steps are performed simultaneously rather than sequentially. [0225] Figure 15 illustrates a heating and cooling jacket 42 in an open position, according to aspects of the present disclosure. As shown in Fig.15, the jacket may include the first shell 46, the second shell 48, the protrusion 102, and may be positioned on the table 44. In some embodiments, the table may include an opening 172 (for example, a circular opening positioned in the center of the top surface of the table 44) such that the magnetic field generator 88 may be disposed therethrough so as to allow for unhindered electromagnetic coupling with the stirrer/impeller 89. In some embodiments, the jacket 42 may include a single hinge 118 (i.e., or multiple hinges 118A, 118B as shown in Fig. 5B). In the embodiment of Fig.15, the first shell 46 is rigidly coupled to the table 44 such that it is concentrically disposed about the opening 172 (i.e., the first shell 46 is positioned so that it is concentric (i.e., radially outward of) half of the circular opening 172; in a closed position, both the first shell 46 and the second shell 48 are concentric about the circulate opening 172). The hinge 118 allows the second shell 48 to swing open and closed such that vessels may be placed within, and removed from, the jacket 42. As shown in Fig. 15, the latch 76 (which may be coupled to the first shell 46) may be used in connection with the catch 170 (positioned on the second shell 48) to receive the latch and keep the jacket 42 secured around the vessel / single use bottle when in operation. Each of the first and second shells 46, 48 may include a plurality of holes 174 positioned within respective circumferential faces 178 (or surfaces 178). The plurality of holes, in some embodiments, act as thermal expansion features. In some embodiments, dowels (not shown) may be positioned in the plurality of holes 174 to help keep the first and second shells 46, 48 positioned relative to each other (for example, via compression fitting of the dowels within the holes 174). The dowels may also help to evenly distribute axial or verticle loading acting on one or both of the first and second shells 46, 48. In some embodiments, the table 44 includes a plurality of feet 176 (for example, four (4) feet that are vertcally adjustable via threading that screws into and out of a bottom surface of the table such that the table may be leveled). [0226] Figure 16 illustrates a heating and cooling jacket 42 in a closed position with the first shell 46 secured to the second shell 48 via latch 76, according to aspects of the present disclosure. The outer casing 180 (for example, a metallic (e.g., stainless steel) outer casing) is visible in Fig. 16. In the embodiment of Fig. 16, the system 150 includes a plurality of inlet tubes 184 disposed through the top of the vessel or bottle, and a plurality of outlet tubes 182 disposed through the elongated through-bore 82, the outlet tubes 182 being in fluid communication with the inlet tubes 184. [0227] Figure 17 illustrates a heating and cooling jacket 42 in operation, according to aspects of the present disclosure. Visible in the embodiment of Fig. 17 are condensation droplets 188 on the outer casing 180 of the jacket 42 (i.e., during a cooling operation). In some embodiments, the jacket 42 and/or system 150 may include a communication wire 186 for communicating signals from the one or more sensors (for example, temperature sensors) that are disposed in the interior of the jacket 42, to the temperature control unit 172 and/or the temperature handling unit 148. The heating and cooling jacket 42 illustrated in Fig. 17 is dimensioned and sized to be used with a 2 L fluid vessel, bioprocessing container, and/or single use bottle. [0228] Figure 18 illustrates a heating and cooling jacket 42 in a closed position without outer casings 180, according to aspects of the present disclosure. In the embodiment of Fig. 18, visible outlet tubing 11890709v1 41
Attorney Docket No.: 2013237-0929 182 is disposed through the elongated through-bore 82, while inlet tubing 184 is disposed through the larger opening 158 at the top of the bottle. [0229] Figs. 19-22 illustrate heating and cooling jackets 42 dimensioned and sized to be used with a 1 L, 5 L, 10 L and 20 L fluid vessels, bioprocessing containers, and/or single use bottles, respectively, according to aspects of the present embodiments. Each of the heating and cooling jackets 42 illustrated in Figs.19-22 are shown with outer casings 180. Table 1 includes various heating and cooling jacket dimensions sized for use with various bottle volumes from 1 L to 50 L. The diameters, heights, and spacings are all in millimeters (mm). As illustrated in Table 1, the jacket diameters (i.e., inner and outer diameters) and heights of the heating and cooling jackets 42 scale up and down approximately in concert with the bottle volume (i.e., the approximate calculated volume that can be derived from the jacket diameter and height scales up and down approximately linearly with the volume of the corresponding bottle that the jacket is configured for). The channel diameters and channel spacings, by contrast, remain within narrow ranges (and do not scale up and down approximately linearly with the bottle volume). For example, as the bottle volume increases from 1 L to 20 L, the channel diameter of the corresponding heating and cooling jacket increases from 12 mm to 15 mm while the channel spacing increases from 7 mm to 10 mm. The number of passes is (i.e., number of vertically stacked channels, each channel extending circumferentially) is a function of the heating and cooling jacket height, the channel diameter and channet spacing. As described herein, the geometry and dimensions of the 50 L heating and cooling jacket 42 are qualitatively different than jackets 42 sized for other volumes, due to manufacturability. Bottle Channel Channel Number of Inner Outer Volume Diameter Spacing Passes Height Diameter Diameter
[0230] Referring still to Table 1, the disclosed dimensions are for exemplary purposes only. Heating and cooling jackets 42 according to the present embodiments may be designed and manufactured using other sizes and dimensions. In some embodiments, heating and cooling jackets 42 according to the present embodiments may include channels 86 with diameters in a narrow range (for example 12mm-15mm, or from about 10mm-18mm, or from about 8mm to about 20 mm) in order to provide enhanced heat transfer (i.e., heating and cooling) from the fluid running through the channels 86 (through the channel walls) to the bioprocessing vessel 52 disposed within the jacket 42. Heating and cooling jackets with channel diameters that are significantly smaller than those of the present embodiments may not allow sufficient mass flow of fluid therethrough to provide adequate heating and/or cooling to the bioprocessing vessel 52. Similarly, heating and cooling jackets with channel diameters that are significantly larger than those of the present embodiments are likely to have reduced cooling and/or heating effectiveness since a larger portion of the thermal fluid flows through the radially outer portion of the channels (i.e., closer to the outer casing and away from the bioprocessing vessel 52, which is where the heating and cooling is needed) thereby resulting in a relative loss of 11890709v1 42
Attorney Docket No.: 2013237-0929 convective heating/cooling to the bioprocessing vessel 52. The smaller channel spacing (for example, 7mm) in the jacket 42 used with the 1 L bottle helps to maximize the number of channel passes 86 that can fit within the smaller height of the 1 L bottle. The larger channel spacing (for example, 10mm) in the jacket 42 used in connection with the 20 L bottle provides additional thickness of the channel walls to help support the larger heating and cooling jacket 42. Accordingly, in some embodiments, the heating and cooling jacket 42 may include a channel diameter to channel spacing ratio that is greater than 1 (for example, that ranges from about 1.2 to about 2.5, or from about 1.4 to about 2.0, or from about 1.5 to about 1.9). [0231] Still referring to Table 1, the 50 L heating and cooling jacket 42, as described herein, may be produced or manufactured via a different from the process than that of the smaller jackets. For example, in some embodiments, heating and cooling jackets 42 sized for 1 L bottles up to and including 20 L jackets may be produced by milling of metal while the 50 L jacket 42 may be turned from a tube. In other words, sheet metal may be rounded to form the cylindrical walls of the 50 L jacket 42 while the planar walls (for example, horizontally oriented and/or radially oriented walls that form and define the channels may be made of flat steel (i.e., sheets of steel) that are machined to share and welded on. Therefore, the cross-sections of the channels of the 50 L jacket 42 may be, for example, rectangular while the cross-sections of the 1 L-20 L jackets 42 may be at least partially circular (for example, semi-circular) as described herein. [0232] Figure 23 illustrates an example of a system including a heating and cooling jacket 42 (and outer casing 180) disposed around a 1 L single use bottle that includes grip features 190, according to aspects of the present disclosure. Figure 24 illustrates an example of a system including grip concavities 192 disposed in a heating and cooling jacket 42, according to aspects of the present disclosure. To accommodate fluid vessels 52 (for example, single use bottles 52 and/or bioreactors 52) that are small enough to carry in one hand (for example, 1 L and 2 L bottles, among other potential sizes) and that include grip features 190, the heating and cooling jackets 42 of the present disclosure may accordingly include corresponding concavites 192 (or as the case may be, convexitites). The concavities 192 and/or convexities 192 mirror the profile of the grip features such that the heating and cooling jacket 42 remains in contact with the single use bottle 52 and/or vessel 52. [0233] Figure 25 illustrates an interior view of a 50 L heating and cooling jacket 42, according to aspects of the present disclosure. Figure 26 illustrates a cross-sectional view of a 50 L heating and cooling jacket 42, according to aspects of the present disclosure. Figure 27 illustrates a perspective view of a 50 L heating and cooling jacket 42, according to aspects of the present disclosure. In the 50 L embodiment(s) shown in Figs. 25-27 (each including both a first shell 46 and a second shell 48), the heating and cooling jacket 42 may include a total of 8 passes or channels including six (6) interior channels 194 and two end channels 196 disposed above and below the interior channels 194. Each of the end channels 196, in some embodiments, include a larger channel height 204 than the channel height 202 of the interior channels 194 (i.e., so as to accommodate and make space for the one or more inlets 66 and/or the one or more outlets 54, which are used for routing cooling and/or heating fluid into and out of the heating and cooling jacket 32). For example, in some embodiments, each of the interior channels 194 includes a first height 202 of about 41 mm (for example, from about 36 mm to about 46 mm, or from about 38 mm to 44 mm) while each of the end channels 196 may include a second height 204 of about 58 mm (for example, from about 53 mm to about 63 mm or from about 55 mm to about 61 mm). Both the interior channels 194 and the end channels 196 may include a width 208 of about 21 mm (for example, from about 16 mm to about 26 mm, or from about 18 mm to about 24 mm). 11890709v1 43
Attorney Docket No.: 2013237-0929 Therefore, in some embodiments, the heating and cooling jacket 42 configured to accommodate 50 L fluid vessels 52 (i.e., single use bottles 52) may include cooling channels with a larger height than width. Cooling channels with a larger channel height 202, 204 than width 208, according to the present disclosure, help to improve cooling and/or heating effectiveness due to the contact surface area between the heating and cooling jacket 42 and the single use bottle 52 / bioreactor 52 being maximized as a ratio of the channel volume. Stated otherwise, taller, thinner cooling channels with an aspect ratio (i.e., height to width ratio) for example, greater than 2, helps to bring the cooling fluid closer to the interior surface of the heating and cooling jacket 42, thereby enhancing the heat transfer effectiveness. In some embodiments, the heating and cooling jacket 42 may include one or more fluid drains 198 (for example, to allow gravity draining of thermal fluids out of the heating and cooling jacket 42) as well as convex contouring 206 disposed in an interior surface of the heating and cooling jacket 42 to match the concave contouring of a single use bottle (or fluid vessel). [0234] Figure 28 illustrates a front view of a mobile bioreactor unit 200 with a heating and cooling jacket 42, according to aspects of the present disclosure. Figure 29 illustrates a side view of a mobile bioreactor unit 200 with a heating and cooling jacket 42, according to aspects of the present disclosure. Figure 30 illustrates a rear view of a mobile bioreactor unit 200 with a heating and cooling jacket 42, according to aspects of the present disclosure. The mobile bioreactor unit 200 of the present disclosure may include a cart 230 with legs (for example, four legs) and wheels 224 coupled to the legs via castors 222 to enhance maneuverability of the mobile bioreactor unit 200. In some embodiments, the mobile bioreactor unit may include a vertically oriented frame 216 for supporting brackets 210 used to support valves (for example, pneumatic valves 234 (shown in Fig.31)) as well as one or more holders 212 for supporting and/or housing a filter capsule 248 (shown in Fig. 31). The cart 230 may be used to support both a 50 L single use bottle 52 / bioreactor 52 (and also smaller size fluid vessels 52) as well as the heating and cooling jacket 42. The cart 230 enables transportability of the mobile bioreactor unit 200. In some embodiments, the mobile bioreactor unit 200 may also include a plurality of supports 94 that extend vertically downward as well as laterally beneath a platform on which the single use bottle / bioreactor 52 and jacket 42 are positioned, for example, to hold a magnetic field generator 88 (shown in Fig. 10). [0235] Referring still to Figs.28-30, (and also to Figs. 33 and 34) mobile bioreactor unit 200 (or system) may include a connection point 220 (for example, a flange 220) for disconnecting the wheels 224 and castors 222 from the legs of the cart 230 such that the cart 230 may be positioned securely on a scale 258 (for example, as shown in Fig. 34). In some embodiments, the cart 230 may include load cells 256 in place of the wheels and castors 224, 222 (as shown in Fig. 33), such that the mobile bioreactor system 200 (including the contents of the single use bottle / bioreactor 52) may be weighed. In some embodiments, the mobile bioreactor system 200 may also include a support plate 218. The support plate 218 may be supported by the frame 216, and itself may be used to support a control cabinet that may contain the control units for: the magnetic stirrer 89 / magnetic field generator 88, one or more pressure sensors, temperature probes and/or valves 234 (shown in Fig. 31). In some embodiments, the single use bottle 52 / bioreactor 52 may also include one or more handles 214 disposed at or near a top end or surface of the single use bottle / bioreactor 52. In some embodiments, the mobile bioreactor system 200 may include a grounding lug 226 to enable electrical grounding of the mobile bioreactor system 200 via a wire connected thereto, to enable use of the mobile bioreactor system 200 with ethanol and other potentially hazardous materials, while maintaining safe use. 11890709v1 44
Attorney Docket No.: 2013237-0929 [0236] Figure 31 illustrates a front view of a mobile bioreactor unit 200 with a heating and cooling jacket, according to aspects of the present disclosure. In some embodiments, the mobile bioreactor unit 200 may include an ethanol supply 242 fluidly coupled upstream of a pump 244 (for example, an ATEX pump) for routing ethanol into the single use bottle 52 / bioreactor 52 via an ethanol supply line 264. A first vent line 260 may be coupled to the ethanol supply line 264 (and may further be connected to a filter (i.e., an EKV filter)) to allow air to be removed from the ethanol supply line 264 upstream of where ethanol enters the single use bottle 52 / bioreactor 52. The mobile bioreactor unit 200 may also include an RNA solution and/or drug substance outlet line 246, for delivering an RNA solution or other biological solution to a downstream process (for example, to a tangential flow filtration module, a t-mixer, a y-mixer, an LNP formation module, a lipoplex formation module, a liposome formation module, another bioreactor, a 0.2 µm filtration module, and/or other types of downstream processes. In some embodiments, the drug substance outlet line 246 may include a filter 248 (i.e., an EKV filter) for removing air from the biological solution via a second vent line 262. The mobile bioreactor unit 200 may also include an air inlet line / air supply line 228 and an air exhaust line / air outlet line 232, each with a valve 234 (i.e., a pneumatic valve) disposed therein, and each being supported by a respective support bracket 210. In some embodiments, each of the air inlet line / air supply line 228 and the air exhaust line / air outlet line 232 may include one or more filters 238, 240 disposed therein. The mobile bioreactor unit 200 may also include: a thermal fluid supply line 254 for routing thermal fluid into the heating and cooling jacket 42 from the temperature control unit (TCU), a TCU return line 252 for routing thermal fluid from the heating and cooling jacket 42 back to the TCU, a magnetic field generator 88 supported by the cart 230, a magnetic stirrer 89 electromagnetically coupled to the magnetic field generator 88 and positioned within the single use bottle 52 / bioreactor 52, and a switch box 250 for electrically coupling the mobile bioreactor unit 200 to an external power supply. [0237] Figure 32 illustrates a perspective view of a mobile bioreactor unit 200 without a heating and cooling jacket, and including the cart 230, grounding lug 226, brackets 210, holders 212, jacket 42, single use bottle / bioreactor 52, and other features, as described herein and according to aspects of the present disclosure. In some embodiments, one of the first shell 46 and the second shell 48 (shown in Figs.25-27) forming the heating and cooling jacket 42 may be rigidly coupled to the planar top surface of the cart 230, for example, via welding, brazing, bolt, screw, nut, glue, epoxy, adhesion, mechanical coupling, and/or other suitable attachment means. Once the single use bottle 52 / bioreactor 52 is placed adjacent the first shell 46 or second shell 48 that is rigidly coupled to the planar top surface of the cart 230, the shell that is not rigidly coupled to the cart 230 may be rotated into place such that the heating and cooling jacket 42 surrounds the single use bottle 52 / bioreactor 52. [0238] In connection with the present disclosed embodiments, the jacket 42 has been described primarily in connection with the use of single use bottles 52. However, the heating and cooling jacket 42 of the present embodiments may be used in connection with any other number of bioprocessing and/or chemical processing vessels including (but not limited to) multi-use or single use bioreactors, tangential flow filtration (TFF) equipment such as TFF cassettes, mixing vessels, reservoirs, other types of filtration devices, thawing chambers, refrigeration units (for example, in place of refrigeration units), and/or in connection with other types of equipment. For example, referring to Fig.2, the heating and cooling jacket 42 of the present embodiments may be used in connection with: one or more bioreactors during in vitro transcription 50; one or more TFF cassettes during a first and/or a second tangential flow filtration process 60, 80; a mixing vessel such 11890709v1 45
Attorney Docket No.: 2013237-0929 as a lipid – RNA impingement jet mixer and/or a lipid-ethanol mixing vessel during LNP formation 70; filtration for bioburden reduction during formulation 90, and/or during in situ storage and/or warehousing (i.e., to provide flexibility in allowing those steps to occur in place rather than having to have separate processes and/or locations for performing those functions). As described herein, the heating and cooling jacket 42 of the present embodiments may be used in connection with with temperature control of an LNP batch, for the cooling of buffers, for cooling in the formulation 90 area, and for cooling in the fill and/finish area 100, as well as in connection with other potential applications. The jacket 42 and bioprocessing vessel 52 may be cylindrical. In some embodiments, the jacket 42 and bioprocessing vessel 52 may be substantially square or cube-shaped with rounded corners. In some embodiments, the bioprocessing vessel 52 may be substantially square or cube shaped with rounded corners while the outer shape of the jacket 42 may be substantially cylindrical, as shown in Figs.9A and 9B. In some embodiments, the bottles / vessels 52 may include a substantially rectangular cross-section, and the jacket 42 may be correspondingly contoured and/or shaped to accommodate the dimensions of the vessel 52. In some embodiments, each of the vessel 52 and jacket 42 may include other shapes. In some embodiments, the heating and cooling jacket 42 may be used in connection with a mobile bioreactor system, as described herein. Equivalents [0239] Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Therefore, the scope of the present invention is not intended to be limited to the above Description. 11890709v1 46
Claims
Attorney Docket No.: 2013237-0929 Claims We claim: 1. A system for heating and/or cooling a vessel, the system comprising: a heating and cooling jacket disposed around the vessel, the jacket comprising a plurality of channels configured to allow a thermal fluid to flow therethrough, the jacket comprising at least one fluid inlet and at least one fluid outlet, each fluidly coupled to the plurality of channels; and a temperature handling unit fluidly coupled to the jacket, the temperature handling unit configured to selectively heat and cool the thermal fluid. 2. The system of claim 1, wherein the fluid comprises at least one of water and silicon oil. 3. The system of claim 1, wherein the vessel comprises at least one of a bioprocessing vessel and a chemical processing vessel. 4. The system of claim 1, wherein the at least one fluid inlet comprises a first fluid inlet and a second fluid inlet, wherein the at least one fluid outlet comprises a first fluid outlet and a second fluid outlet, the first fluid outlet being fluidly coupled to the first fluid inlet via a first fluid stream within the jacket, and the second fluid outlet being fluidly coupled to the second fluid inlet via a second fluid stream within the jacket, and wherein the first fluid stream and the second fluid stream are fluidly disconnected within the jacket. 5. The system of claim 1, further comprising: at least one pump disposed fluidly upstream of the temperature handing unit; and at least one reservoir disposed fluidly upstream of the at least one pump. 6. The system of claim 5, further comprising a temperature control unit (TCU) communicatively coupled to both the temperature handling unit and the at least one pump. 7. The system of claim 1, further comprising at least one support table for supporting the vessel and the jacket, wherein the at least one support table comprises at least one of a magnetic field generator and an inducer configured to be magnetically coupled to, and to cause rotation of, at least one of an impeller and a stirrer disposed within the vessel. 8. The system of claim 3, wherein the vessel comprises a bioprocessing vessel comprising at last one of a bioreactor and a single use bottle. 9. The system of claim 8, wherein the vessel comprises at least one concavity. 11890709v1 47
Attorney Docket No.: 2013237-0929 10. The system of claim 9, wherein the jacket comprises at least one protrusion comprising contouring that corresponds to the at least one concavity of the vessel, the at least one protrusion protruding radially inward of a nominal inner diameter of the jacket. 11. A heating and cooling jacket comprising: a first semi-cylindrical shell comprising a first plurality of channels configured to allow a thermal fluid to flow therethrough, the first shell comprising a first fluid inlet and a first fluid outlet, each fluidly coupled to the first plurality of channels; and a second semi-cylindrical shell comprising a second plurality of channels configured to allow the thermal fluid to flow therethrough, the second shell comprising a second fluid inlet and a second fluid outlet, each fluidly coupled to the second plurality of channels; wherein the first and second shells form the jacket and are configured to both heat and cool, via the thermal fluid, at least one object disposed within the jacket. 12. The jacket of claim 11, comprising at least one hinge coupling the first shell to the second shell, wherein in a closed position, the jacket comprises a substantially cylindrical shape. 13. The jacket of claim 11, wherein each of the first and second pluralities of channels comprises a plurality of adjacent channels, each of which extends circumferentially around the respective first or second shell, and wherein each of the channels comprises a substantially semicircular cross-section. 14. The jacket of claim 13, wherein each of the first shell and the second shell comprises an outer casing that defines a radially outer wall of the channels, and wherein the outer casings are composed of at least one structural polymer material. 15. The jacket of claim 11, wherein each of the first and second shells are composed of stainless steel. 16. The jacket of claim 11, wherein each of the first and second pluralities of channels comprise channels with a uniform outer diameter; and wherein each of the first and second pluralities of channels comprise at least one channel that is positioned at a different inner diameter than at least one other channel within the same shell. 17. The jacket of claim 11, wherein at least one of the first shell and the second shell comprises at least one through-bore that connects an interior of the shell to an exterior of the shell. 18. A method of heating or cooling an object comprising: providing a cooling and heating jacket comprising a plurality of channels configured to allow flow of a thermal fluid therethrough; positioning at least one object to be heated or cooled within the jacket; circulating the thermal fluid through the plurality of channels such that the object is heated or cooled to a first temperature for a first period of time; and 11890709v1 48
Attorney Docket No.: 2013237-0929 circulating the thermal fluid through the plurality of channels such that the object is heated or cooled to a second temperature for a second period of time; wherein at least one of the first temperature and the second temperature is below room temperature, and wherein at least one of first temperature and the second temperature is above room temperature. 19. The method of claim 18, wherein there is at least a 15 deg C difference between the first temperature and the second temperature, wherein at least one of the first temperature and the second temperature is in a range from about 1 deg C to about 17 deg C, and wherein at least one of the first temperature and the second temperature is in a range from about 26 deg C to about 40 deg C. 20. The method of claim 18, wherein the jacket is configured to heat and cool the object to temperatures in a range from about 1 deg C to about 99 deg C. 21. The system of claim 1, wherein the vessel comprises at least one of: (1) one or more bioreactors being used for in vitro transcription, (2) one or more TFF cassettes being used for a first and/or a second tangential flow filtration process, (3) one or more mixing vessels containing a mix of lipids and RNA solution, and/or (4) a lipid-ethanol mixing vessel being used for an LNP formation process. 22. The method of claim 18, wherein the thermal fluid comprises at least one of a water-glycol mixture and a water-ethylene-glycol mixture, wherein the thermal fluid comprises a pH in a range from about 6.0 to about 8.5, and wherein a density of the thermal fluid does not exceed 1kg/dm^3. 23. The system of claim 1, wherein the thermal fluid comprises at least one of a water-glycol mixture and a water-ethylene-glycol mixture, wherein the thermal fluid comprises a pH in a range from about 6.0 to about 8.5, and wherein a density of the thermal fluid does not exceed 1kg/dm^3. 24. The system of claim 1, wherein the thermal fluid comprises a water-glycol mixture, and wherein cooling and heating of the heating and cooling jacket is limited to a temperature range from about 2 deg C to about 35 deg C. 25. The system of claim 1, wherein the heating and cooling jacket disposed around the vessel accommodates a vessel volume of at least one of 1 L, 2 L, 5 L, 10 L, 20 L, and 50 L. 26. The system of claim 1, wherein the vessel comprises a volume of at least one of 1 L, 2 L, 5 L, 10 L, 20 L, and 50 L. 27. The system of claim 1, wherein each channel of the plurality of channels comprises a nominal diameter in a range from about 8 mm to about 20 mm. 11890709v1 49
Attorney Docket No.: 2013237-0929 28. The system of claim 1, wherein the heating and cooling jacket comprises a plurality of vertical spaces vertically separating each channel of the plurality of channels from a channel immediately above and/or below each channel. 29. The system of claim 28, wherein each vertical space of the plurality of vertical spaces comprises a height of from about 7 mm to about 10 mm. 30. The system of claim 1, wherein the heating and cooling jacket comprises a substantially cylindrical shape; wherein each channel of the plurality of channels extends circumferentially within the heating and cooling jacket around at least a portion of the circumference of the heating and cooling jacket; and wherein each channel of the plurality of channels is disposed above and/or below one or more adjacent channels of the plurality of channels. 31. The system of claim 30, comprising at least one continuous fluid path through the heating and cooling jacket, the at least one continuous fluid path comprising each channel of the plurality of channels, wherein the plurality of channels comprises from about 5 to about 20 channels. 32. The system of claim 31, wherein the at least one continuous fluid path comprises a plurality of connections disposed at circumferential end(s) of each channel of the plurality of channels, wherein each connection of the plurality of connections connects a channel of the plurality of channels to at least one other channel of the plurality of channels. 33. The system of claim 32, wherein each connection of the plurality of connections causes the at least one continuous fluid path to make a 180-degree turn. 34. The system of claim 31, wherein the heating and cooling jacket comprises a first semi-cylindrical shell and a second semi-cylindrical shell, wherein the at least one continuous fluid path comprises at least two continuous fluid paths disposed through the heating and cooling jacket, and wherein the at least two continuous fluid paths comprise a first continuous fluid path disposed through the first semi-cylindrical shell and a second continuous fluid path disposed through the second semi-cylindrical shell. 35. The system of claim 1, wherein the heating and cooling jacket comprises a height in a range from about 100 mm to about 600 mm. 36. The system of claim 1, wherein the heating and cooling jacket comprises a height in a range from about 230 mm to about 300 mm. 11890709v1 50
Attorney Docket No.: 2013237-0929 37. The system of claim 1, wherein the heating and cooling jacket comprises an outer diameter in a range from about 100 mm to about 500 mm. 38. The system of claim 1, wherein the heating and cooling jacket comprises an outer diameter in a range from about 230 mm to about 330 mm. 39. The system of claim 1, wherein the heating and cooling jacket comprises a height in a range from about 290 mm to about 390 mm. 40. The system of claim 27, wherein each channel of the plurality of channels comprises a nominal diameter in a range from about 12 mm to about 15 mm. 41. The system of claim 28, wherein the heating and cooling jacket comprises material forming the walls of each channel disposed within each vertical space of the plurality of vertical spaces. 42. The system of claim 28, wherein the heating and cooling jacket comprises a channel diameter to channel spacing ratio in a range from about 1.2 to about 2.5. 43. The system of claim 28, wherein the heating and cooling jacket comprises a channel diameter to channel spacing ratio in a range from about 1.4 to about 1.9. 44. The jacket of claim 11, comprising at least one sensor access port disposed in at least one of the first shell and the second shell, the at least one sensor access port comprising a first rounded portion, a second rounded portion, and an elongated portion disposed between the first rounded portion and the second rounded portion. 45. A mobile bioreactor system comprising: a movable cart; a bioreactor disposed on the moveable cart; and the jacket of any of the preceding claims, the jacket being disposed around the bioreactor. 46. The system of claim 45, wherein the jacket comprises a first shell hingedly connected to a second shell, and wherein one of the first shell and the second shell is rigidly coupled to a top surface of the movable cart. 47. The system of claim 45, wherein the movable cart comprises wheels and castors, and wherein the wheels and castors are removably coupled to the cart. 48. The system of claim 45, wherein the bioreactor comprises a nominal volume of 50 L. 11890709v1 51
Attorney Docket No.: 2013237-0929 49. The system of claim 45, wherein the movable cart comprises a vertically oriented frame for supporting at least one of a bracket, a filter holder, a support plate, and an electrical switchbox. 50. The system of claim 45, wherein the jacket comprises channels with an aspect ratio (height to width ratio) of greater than 2.0. 11890709v1 52
Applications Claiming Priority (4)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US202363458019P | 2023-04-07 | 2023-04-07 | |
US63/458,019 | 2023-04-07 | ||
US202363464990P | 2023-05-09 | 2023-05-09 | |
US63/464,990 | 2023-05-09 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2024209418A1 true WO2024209418A1 (en) | 2024-10-10 |
Family
ID=90829357
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/IB2024/053349 WO2024209418A1 (en) | 2023-04-07 | 2024-04-05 | Heating and cooling jacket for fluid vessels |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2024209418A1 (en) |
Citations (10)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20040062140A1 (en) * | 2002-09-27 | 2004-04-01 | Cadogan David Phillip | Bioprocess container, bioprocess container mixing device and method of use thereof |
US8058069B2 (en) | 2008-04-15 | 2011-11-15 | Protiva Biotherapeutics, Inc. | Lipid formulations for nucleic acid delivery |
US20130089925A1 (en) * | 2008-03-25 | 2013-04-11 | Xcellerex, Inc. | Temperature controlling surfaces and support structures |
CZ2014441A3 (en) * | 2014-06-27 | 2016-01-06 | Tomáš Pink | Processing container |
US20160184827A1 (en) * | 2013-07-31 | 2016-06-30 | Sartorius Stedim Biotech Gmbh | Temperature control device, use and arrangement |
US20160208209A1 (en) * | 2015-01-19 | 2016-07-21 | Dakota Systems, Inc. | Bioreactor Docking Station Systems, and Methods of Use Thereof |
WO2017075531A1 (en) | 2015-10-28 | 2017-05-04 | Acuitas Therapeutics, Inc. | Novel lipids and lipid nanoparticle formulations for delivery of nucleic acids |
WO2018081480A1 (en) | 2016-10-26 | 2018-05-03 | Acuitas Therapeutics, Inc. | Lipid nanoparticle formulations |
US20210261897A1 (en) * | 2018-06-28 | 2021-08-26 | Curevac Ag | Bioreactor for rna in vitro transcription |
US20230025193A1 (en) * | 2019-12-23 | 2023-01-26 | Global Life Sciences Solutions Usa Llc | System having a tube shaft impeller and an associated method thereof |
-
2024
- 2024-04-05 WO PCT/IB2024/053349 patent/WO2024209418A1/en unknown
Patent Citations (10)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20040062140A1 (en) * | 2002-09-27 | 2004-04-01 | Cadogan David Phillip | Bioprocess container, bioprocess container mixing device and method of use thereof |
US20130089925A1 (en) * | 2008-03-25 | 2013-04-11 | Xcellerex, Inc. | Temperature controlling surfaces and support structures |
US8058069B2 (en) | 2008-04-15 | 2011-11-15 | Protiva Biotherapeutics, Inc. | Lipid formulations for nucleic acid delivery |
US20160184827A1 (en) * | 2013-07-31 | 2016-06-30 | Sartorius Stedim Biotech Gmbh | Temperature control device, use and arrangement |
CZ2014441A3 (en) * | 2014-06-27 | 2016-01-06 | Tomáš Pink | Processing container |
US20160208209A1 (en) * | 2015-01-19 | 2016-07-21 | Dakota Systems, Inc. | Bioreactor Docking Station Systems, and Methods of Use Thereof |
WO2017075531A1 (en) | 2015-10-28 | 2017-05-04 | Acuitas Therapeutics, Inc. | Novel lipids and lipid nanoparticle formulations for delivery of nucleic acids |
WO2018081480A1 (en) | 2016-10-26 | 2018-05-03 | Acuitas Therapeutics, Inc. | Lipid nanoparticle formulations |
US20210261897A1 (en) * | 2018-06-28 | 2021-08-26 | Curevac Ag | Bioreactor for rna in vitro transcription |
US20230025193A1 (en) * | 2019-12-23 | 2023-01-26 | Global Life Sciences Solutions Usa Llc | System having a tube shaft impeller and an associated method thereof |
Non-Patent Citations (8)
Title |
---|
"Making and Using Antibodies: A Practical Handbook", 29 July 2013, CRC PRESS |
"Monoclonal Antibodies: Methods and Protocols (Methods in Molecular Biology).", 12 February 2014, HUMANA PRESS |
CULLIS ET AL., MOLECULAR THERAPY, vol. 25, 5 July 2017 (2017-07-05), pages 1467 |
GREENSAMBROOK: "Molecular Cloning: A Laboratory Manual", 2012, COLD SPRING HARBOR LABORATORY PRESS |
HOU ET AL., NAT. REV. MATER, 10 August 2021 (2021-08-10), Retrieved from the Internet <URL:https://doi.org/10.1038/s41578-021-00358-0> |
MEYERSMILLER, CABIOS, vol. 4, 1989, pages 11 - 17 |
MICHAEL STEINITZ: "Human Monoclonal Antibodies: Methods and Protocols (Methods in Molecular Biology).", 30 September 2013, COLD SPRING HARBOR LABORATORY PRESS |
PATRICK CHAMES: "Antibody Engineering: Methods and Protocols", 21 August 2012, HUMANA PRESS |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
Fragasso et al. | Reconstitution of ultrawide DNA origami pores in liposomes for transmembrane transport of macromolecules | |
JP2022543423A (en) | Manufacturing methods and devices for removing materials from therapeutic compositions | |
Wagner et al. | Liposome technology for industrial purposes | |
US20210023514A1 (en) | Continuous flow systems with bifurcating mixers | |
BR112020025601A2 (en) | bioreactor for in vitro transcription of rna | |
US20210128474A1 (en) | Methods for manufacturing a liposome encapsulated rna | |
CN103167866A (en) | Novel low molecular weight cationic lipids for oligonucleotide delivery | |
Liu et al. | Design strategies for and stability of mRNA–lipid nanoparticle COVID-19 vaccines | |
Choi et al. | Targeted delivery of exosomes armed with anti-cancer therapeutics | |
US20240246056A1 (en) | Manufacturing device for a pharmaceutical product | |
WO2024209418A1 (en) | Heating and cooling jacket for fluid vessels | |
Tanaka et al. | Ready-to-use-type lyophilized lipid nanoparticle formulation for the postencapsulation of messenger RNA | |
Latulippe et al. | Elongational flow model for transmission of supercoiled plasmid DNA during membrane ultrafiltration | |
US20240110214A1 (en) | Rna manufacturing | |
IL302970A (en) | A device for preparing a dna product by means of capillary polymerase chain reaction | |
WO2024194767A1 (en) | Automated drug production system | |
US20230201817A1 (en) | Modular drug production system | |
Lu et al. | Regulatory perspective for quality evaluation of lipid nanoparticle-based mRNA vaccines in China | |
WO2024083345A1 (en) | Methods and uses associated with liquid compositions | |
NL2027654B1 (en) | Methods for manufacturing a syntetic template | |
CN116096485A (en) | Flow channel structure, method for stirring fluid, and method for producing lipid particles | |
EP4378573A1 (en) | Chip for preparing lipid nanoparticles, system for preparing lipid nanoparticles comprising same, and method for preparing lipid nanoparticles | |
US20230332088A1 (en) | Methods, apparatus and products of cell, tissue engineering and vaccine/antibody production systems | |
Trollmann et al. | Mechanistic Insight into pH-Driven Phase Transition of Lipid Nanoparticles | |
CN118871097A (en) | Nucleic acid lipid nanoparticle with low empty rate and preparation method thereof |