WO2023019213A1 - Three-dimensional culturing of pluripotent stem cells to produce hematopoietic stem cells - Google Patents
Three-dimensional culturing of pluripotent stem cells to produce hematopoietic stem cells Download PDFInfo
- Publication number
- WO2023019213A1 WO2023019213A1 PCT/US2022/074853 US2022074853W WO2023019213A1 WO 2023019213 A1 WO2023019213 A1 WO 2023019213A1 US 2022074853 W US2022074853 W US 2022074853W WO 2023019213 A1 WO2023019213 A1 WO 2023019213A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- cells
- stem cells
- cell
- bioreactor
- concentration
- Prior art date
Links
- 210000001778 pluripotent stem cell Anatomy 0.000 title claims abstract description 33
- 210000003958 hematopoietic stem cell Anatomy 0.000 title claims abstract description 25
- 238000012258 culturing Methods 0.000 title claims description 15
- 210000000822 natural killer cell Anatomy 0.000 claims abstract description 99
- 108010019670 Chimeric Antigen Receptors Proteins 0.000 claims abstract description 26
- 210000004263 induced pluripotent stem cell Anatomy 0.000 claims abstract description 22
- 210000001671 embryonic stem cell Anatomy 0.000 claims abstract description 15
- 210000004027 cell Anatomy 0.000 claims description 181
- 238000000034 method Methods 0.000 claims description 75
- 239000006143 cell culture medium Substances 0.000 claims description 21
- 108010049955 Bone Morphogenetic Protein 4 Proteins 0.000 claims description 14
- 102100024505 Bone morphogenetic protein 4 Human genes 0.000 claims description 14
- 239000013598 vector Substances 0.000 claims description 14
- 101000581981 Homo sapiens Neural cell adhesion molecule 1 Proteins 0.000 claims description 13
- 102100027347 Neural cell adhesion molecule 1 Human genes 0.000 claims description 13
- 150000007523 nucleic acids Chemical group 0.000 claims description 13
- 210000000130 stem cell Anatomy 0.000 claims description 12
- 102100031573 Hematopoietic progenitor cell antigen CD34 Human genes 0.000 claims description 11
- 101000777663 Homo sapiens Hematopoietic progenitor cell antigen CD34 Proteins 0.000 claims description 11
- CIWBSHSKHKDKBQ-JLAZNSOCSA-N Ascorbic acid Chemical compound OC[C@H](O)[C@H]1OC(=O)C(O)=C1O CIWBSHSKHKDKBQ-JLAZNSOCSA-N 0.000 claims description 10
- 108010073929 Vascular Endothelial Growth Factor A Proteins 0.000 claims description 10
- 102000005789 Vascular Endothelial Growth Factors Human genes 0.000 claims description 10
- 108010019530 Vascular Endothelial Growth Factors Proteins 0.000 claims description 10
- 108091028043 Nucleic acid sequence Proteins 0.000 claims description 9
- 108700014844 flt3 ligand Proteins 0.000 claims description 9
- 239000001963 growth medium Substances 0.000 claims description 9
- HZAXFHJVJLSVMW-UHFFFAOYSA-N 2-Aminoethan-1-ol Chemical compound NCCO HZAXFHJVJLSVMW-UHFFFAOYSA-N 0.000 claims description 8
- 102000003812 Interleukin-15 Human genes 0.000 claims description 8
- 108090000172 Interleukin-15 Proteins 0.000 claims description 8
- 239000000427 antigen Substances 0.000 claims description 8
- 102000036639 antigens Human genes 0.000 claims description 8
- 108091007433 antigens Proteins 0.000 claims description 8
- DGVVWUTYPXICAM-UHFFFAOYSA-N β‐Mercaptoethanol Chemical compound OCCS DGVVWUTYPXICAM-UHFFFAOYSA-N 0.000 claims description 8
- 108010002386 Interleukin-3 Proteins 0.000 claims description 7
- 102000000646 Interleukin-3 Human genes 0.000 claims description 7
- 108010002586 Interleukin-7 Proteins 0.000 claims description 7
- 102000000704 Interleukin-7 Human genes 0.000 claims description 7
- 108010056030 retronectin Proteins 0.000 claims description 7
- IDDDVXIUIXWAGJ-DDSAHXNVSA-N 4-[(1r)-1-aminoethyl]-n-pyridin-4-ylcyclohexane-1-carboxamide;dihydrochloride Chemical group Cl.Cl.C1CC([C@H](N)C)CCC1C(=O)NC1=CC=NC=C1 IDDDVXIUIXWAGJ-DDSAHXNVSA-N 0.000 claims description 6
- 229960005070 ascorbic acid Drugs 0.000 claims description 5
- 235000010323 ascorbic acid Nutrition 0.000 claims description 5
- 239000011668 ascorbic acid Substances 0.000 claims description 5
- BVTBRVFYZUCAKH-UHFFFAOYSA-L disodium selenite Chemical compound [Na+].[Na+].[O-][Se]([O-])=O BVTBRVFYZUCAKH-UHFFFAOYSA-L 0.000 claims description 5
- 239000003112 inhibitor Substances 0.000 claims description 5
- 229940076264 interleukin-3 Drugs 0.000 claims description 5
- 229940100994 interleukin-7 Drugs 0.000 claims description 5
- 239000011435 rock Substances 0.000 claims description 5
- 229960001471 sodium selenite Drugs 0.000 claims description 5
- 235000015921 sodium selenite Nutrition 0.000 claims description 5
- 239000011781 sodium selenite Substances 0.000 claims description 5
- 108010088751 Albumins Proteins 0.000 claims description 4
- 102000009027 Albumins Human genes 0.000 claims description 4
- 239000004372 Polyvinyl alcohol Substances 0.000 claims description 4
- 230000004913 activation Effects 0.000 claims description 4
- 239000000556 agonist Substances 0.000 claims description 4
- 239000012634 fragment Substances 0.000 claims description 4
- 230000003834 intracellular effect Effects 0.000 claims description 4
- 150000002632 lipids Chemical class 0.000 claims description 4
- 229920002451 polyvinyl alcohol Polymers 0.000 claims description 4
- 235000019422 polyvinyl alcohol Nutrition 0.000 claims description 4
- 230000002463 transducing effect Effects 0.000 claims description 3
- 102000015696 Interleukins Human genes 0.000 claims description 2
- 108010063738 Interleukins Proteins 0.000 claims description 2
- 238000004519 manufacturing process Methods 0.000 claims description 2
- 102000000568 rho-Associated Kinases Human genes 0.000 claims description 2
- 108010041788 rho-Associated Kinases Proteins 0.000 claims description 2
- 230000004069 differentiation Effects 0.000 abstract description 55
- 238000004113 cell culture Methods 0.000 abstract description 10
- 210000002540 macrophage Anatomy 0.000 abstract 1
- 239000002609 medium Substances 0.000 description 42
- 230000014509 gene expression Effects 0.000 description 32
- 206010028980 Neoplasm Diseases 0.000 description 31
- 238000010361 transduction Methods 0.000 description 28
- 230000026683 transduction Effects 0.000 description 28
- 230000002147 killing effect Effects 0.000 description 27
- 201000011510 cancer Diseases 0.000 description 26
- 238000000338 in vitro Methods 0.000 description 18
- 108010005327 CD19-specific chimeric antigen receptor Proteins 0.000 description 16
- 241000713666 Lentivirus Species 0.000 description 14
- 230000006698 induction Effects 0.000 description 13
- 230000024245 cell differentiation Effects 0.000 description 12
- 238000001727 in vivo Methods 0.000 description 12
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 description 11
- 238000000684 flow cytometry Methods 0.000 description 11
- 230000003394 haemopoietic effect Effects 0.000 description 11
- 239000002356 single layer Substances 0.000 description 11
- 230000006978 adaptation Effects 0.000 description 10
- 238000012239 gene modification Methods 0.000 description 10
- 230000005017 genetic modification Effects 0.000 description 10
- 235000013617 genetically modified food Nutrition 0.000 description 10
- 239000003550 marker Substances 0.000 description 10
- 101000738771 Homo sapiens Receptor-type tyrosine-protein phosphatase C Proteins 0.000 description 9
- 102100037422 Receptor-type tyrosine-protein phosphatase C Human genes 0.000 description 9
- 238000003556 assay Methods 0.000 description 9
- 230000003013 cytotoxicity Effects 0.000 description 8
- 231100000135 cytotoxicity Toxicity 0.000 description 8
- 210000001939 mature NK cell Anatomy 0.000 description 8
- 230000001177 retroviral effect Effects 0.000 description 8
- 108020004414 DNA Proteins 0.000 description 7
- 241000699666 Mus <mouse, genus> Species 0.000 description 7
- 238000004458 analytical method Methods 0.000 description 7
- 230000010261 cell growth Effects 0.000 description 7
- 238000001802 infusion Methods 0.000 description 7
- 230000000977 initiatory effect Effects 0.000 description 7
- 230000002688 persistence Effects 0.000 description 7
- 239000006228 supernatant Substances 0.000 description 7
- HJCMDXDYPOUFDY-WHFBIAKZSA-N Ala-Gln Chemical compound C[C@H](N)C(=O)N[C@H](C(O)=O)CCC(N)=O HJCMDXDYPOUFDY-WHFBIAKZSA-N 0.000 description 6
- 102000004127 Cytokines Human genes 0.000 description 6
- 108090000695 Cytokines Proteins 0.000 description 6
- 238000011161 development Methods 0.000 description 6
- 230000018109 developmental process Effects 0.000 description 6
- 238000005516 engineering process Methods 0.000 description 6
- 238000009169 immunotherapy Methods 0.000 description 6
- 239000000203 mixture Substances 0.000 description 6
- 238000004264 monolayer culture Methods 0.000 description 6
- 108020004707 nucleic acids Proteins 0.000 description 6
- 102000039446 nucleic acids Human genes 0.000 description 6
- 241001430294 unidentified retrovirus Species 0.000 description 6
- 101001109501 Homo sapiens NKG2-D type II integral membrane protein Proteins 0.000 description 5
- 101000589305 Homo sapiens Natural cytotoxicity triggering receptor 2 Proteins 0.000 description 5
- 101000971513 Homo sapiens Natural killer cells antigen CD94 Proteins 0.000 description 5
- 108060001084 Luciferase Proteins 0.000 description 5
- 239000005089 Luciferase Substances 0.000 description 5
- 241000699670 Mus sp. Species 0.000 description 5
- 102100022680 NKG2-D type II integral membrane protein Human genes 0.000 description 5
- 102100032851 Natural cytotoxicity triggering receptor 2 Human genes 0.000 description 5
- 102100021462 Natural killer cells antigen CD94 Human genes 0.000 description 5
- 241000700605 Viruses Species 0.000 description 5
- 210000002242 embryoid body Anatomy 0.000 description 5
- 230000003389 potentiating effect Effects 0.000 description 5
- 230000008569 process Effects 0.000 description 5
- 238000011002 quantification Methods 0.000 description 5
- 210000002966 serum Anatomy 0.000 description 5
- 230000009469 supplementation Effects 0.000 description 5
- 101000600434 Homo sapiens Putative uncharacterized protein encoded by MIR7-3HG Proteins 0.000 description 4
- 102100037401 Putative uncharacterized protein encoded by MIR7-3HG Human genes 0.000 description 4
- 102100035140 Vitronectin Human genes 0.000 description 4
- 108010031318 Vitronectin Proteins 0.000 description 4
- 238000013459 approach Methods 0.000 description 4
- 230000008901 benefit Effects 0.000 description 4
- 230000015572 biosynthetic process Effects 0.000 description 4
- 238000002619 cancer immunotherapy Methods 0.000 description 4
- 238000003501 co-culture Methods 0.000 description 4
- 210000004700 fetal blood Anatomy 0.000 description 4
- 230000012010 growth Effects 0.000 description 4
- 238000002347 injection Methods 0.000 description 4
- 239000007924 injection Substances 0.000 description 4
- 230000035800 maturation Effects 0.000 description 4
- -1 phosphorotriesters Chemical class 0.000 description 4
- 230000002062 proliferating effect Effects 0.000 description 4
- 230000035755 proliferation Effects 0.000 description 4
- RXWNCPJZOCPEPQ-NVWDDTSBSA-N puromycin Chemical compound C1=CC(OC)=CC=C1C[C@H](N)C(=O)N[C@H]1[C@@H](O)[C@H](N2C3=NC=NC(=C3N=C2)N(C)C)O[C@@H]1CO RXWNCPJZOCPEPQ-NVWDDTSBSA-N 0.000 description 4
- 239000000725 suspension Substances 0.000 description 4
- 102100024222 B-lymphocyte antigen CD19 Human genes 0.000 description 3
- 101000980825 Homo sapiens B-lymphocyte antigen CD19 Proteins 0.000 description 3
- 108010076089 accutase Proteins 0.000 description 3
- 230000001464 adherent effect Effects 0.000 description 3
- 230000005907 cancer growth Effects 0.000 description 3
- 230000022534 cell killing Effects 0.000 description 3
- 238000012512 characterization method Methods 0.000 description 3
- 230000002596 correlated effect Effects 0.000 description 3
- 238000005138 cryopreservation Methods 0.000 description 3
- 239000012636 effector Substances 0.000 description 3
- 239000012091 fetal bovine serum Substances 0.000 description 3
- 238000010230 functional analysis Methods 0.000 description 3
- 238000003384 imaging method Methods 0.000 description 3
- 239000002773 nucleotide Substances 0.000 description 3
- 125000003729 nucleotide group Chemical group 0.000 description 3
- 239000008188 pellet Substances 0.000 description 3
- 239000000047 product Substances 0.000 description 3
- 108090000623 proteins and genes Proteins 0.000 description 3
- 230000035899 viability Effects 0.000 description 3
- 108091026890 Coding region Proteins 0.000 description 2
- 102000053602 DNA Human genes 0.000 description 2
- 102100020715 Fms-related tyrosine kinase 3 ligand protein Human genes 0.000 description 2
- 101710162577 Fms-related tyrosine kinase 3 ligand protein Proteins 0.000 description 2
- 102100035423 POU domain, class 5, transcription factor 1 Human genes 0.000 description 2
- 101710126211 POU domain, class 5, transcription factor 1 Proteins 0.000 description 2
- 108091093037 Peptide nucleic acid Proteins 0.000 description 2
- 108091028664 Ribonucleotide Proteins 0.000 description 2
- UIIMBOGNXHQVGW-UHFFFAOYSA-M Sodium bicarbonate Chemical compound [Na+].OC([O-])=O UIIMBOGNXHQVGW-UHFFFAOYSA-M 0.000 description 2
- 238000013019 agitation Methods 0.000 description 2
- 230000000259 anti-tumor effect Effects 0.000 description 2
- 239000007640 basal medium Substances 0.000 description 2
- 239000008280 blood Substances 0.000 description 2
- 239000006285 cell suspension Substances 0.000 description 2
- 230000008859 change Effects 0.000 description 2
- 230000000052 comparative effect Effects 0.000 description 2
- 150000001875 compounds Chemical class 0.000 description 2
- 239000003814 drug Substances 0.000 description 2
- 230000000694 effects Effects 0.000 description 2
- 238000012632 fluorescent imaging Methods 0.000 description 2
- 238000003306 harvesting Methods 0.000 description 2
- 238000013537 high throughput screening Methods 0.000 description 2
- NOESYZHRGYRDHS-UHFFFAOYSA-N insulin Chemical compound N1C(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(NC(=O)CN)C(C)CC)CSSCC(C(NC(CO)C(=O)NC(CC(C)C)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CCC(N)=O)C(=O)NC(CC(C)C)C(=O)NC(CCC(O)=O)C(=O)NC(CC(N)=O)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CSSCC(NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2C=CC(O)=CC=2)NC(=O)C(CC(C)C)NC(=O)C(C)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2NC=NC=2)NC(=O)C(CO)NC(=O)CNC2=O)C(=O)NCC(=O)NC(CCC(O)=O)C(=O)NC(CCCNC(N)=N)C(=O)NCC(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC(O)=CC=3)C(=O)NC(C(C)O)C(=O)N3C(CCC3)C(=O)NC(CCCCN)C(=O)NC(C)C(O)=O)C(=O)NC(CC(N)=O)C(O)=O)=O)NC(=O)C(C(C)CC)NC(=O)C(CO)NC(=O)C(C(C)O)NC(=O)C1CSSCC2NC(=O)C(CC(C)C)NC(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CC(N)=O)NC(=O)C(NC(=O)C(N)CC=1C=CC=CC=1)C(C)C)CC1=CN=CN1 NOESYZHRGYRDHS-UHFFFAOYSA-N 0.000 description 2
- 238000007912 intraperitoneal administration Methods 0.000 description 2
- 210000004698 lymphocyte Anatomy 0.000 description 2
- 238000012423 maintenance Methods 0.000 description 2
- 239000000463 material Substances 0.000 description 2
- 230000004048 modification Effects 0.000 description 2
- 238000012986 modification Methods 0.000 description 2
- 238000010172 mouse model Methods 0.000 description 2
- 235000015097 nutrients Nutrition 0.000 description 2
- 210000005259 peripheral blood Anatomy 0.000 description 2
- 239000011886 peripheral blood Substances 0.000 description 2
- 230000002085 persistent effect Effects 0.000 description 2
- 230000002028 premature Effects 0.000 description 2
- 238000002360 preparation method Methods 0.000 description 2
- 229950010131 puromycin Drugs 0.000 description 2
- 230000009467 reduction Effects 0.000 description 2
- 230000001105 regulatory effect Effects 0.000 description 2
- 239000002336 ribonucleotide Substances 0.000 description 2
- 229920002477 rna polymer Polymers 0.000 description 2
- 238000013341 scale-up Methods 0.000 description 2
- 238000003756 stirring Methods 0.000 description 2
- UCSJYZPVAKXKNQ-HZYVHMACSA-N streptomycin Chemical compound CN[C@H]1[C@H](O)[C@@H](O)[C@H](CO)O[C@H]1O[C@@H]1[C@](C=O)(O)[C@H](C)O[C@H]1O[C@@H]1[C@@H](NC(N)=N)[C@H](O)[C@@H](NC(N)=N)[C@H](O)[C@H]1O UCSJYZPVAKXKNQ-HZYVHMACSA-N 0.000 description 2
- 229960005322 streptomycin Drugs 0.000 description 2
- 210000002536 stromal cell Anatomy 0.000 description 2
- 239000000758 substrate Substances 0.000 description 2
- 238000004114 suspension culture Methods 0.000 description 2
- 230000002123 temporal effect Effects 0.000 description 2
- 238000012546 transfer Methods 0.000 description 2
- 238000013519 translation Methods 0.000 description 2
- 238000011282 treatment Methods 0.000 description 2
- 210000003462 vein Anatomy 0.000 description 2
- 230000003612 virological effect Effects 0.000 description 2
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 1
- QPXGLRJFCBZQNF-UHFFFAOYSA-N 1-[3-(hydroxymethyl)phenyl]-2-[(2-phenylmethoxyphenyl)methylamino]ethanol Chemical compound OCC1=CC=CC(C(O)CNCC=2C(=CC=CC=2)OCC=2C=CC=CC=2)=C1 QPXGLRJFCBZQNF-UHFFFAOYSA-N 0.000 description 1
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 1
- 102000017420 CD3 protein, epsilon/gamma/delta subunit Human genes 0.000 description 1
- 102000003974 Fibroblast growth factor 2 Human genes 0.000 description 1
- 108090000379 Fibroblast growth factor 2 Proteins 0.000 description 1
- 108090000288 Glycoproteins Proteins 0.000 description 1
- 102000003886 Glycoproteins Human genes 0.000 description 1
- 101001027128 Homo sapiens Fibronectin Proteins 0.000 description 1
- 101001055157 Homo sapiens Interleukin-15 Proteins 0.000 description 1
- 101000738335 Homo sapiens T-cell surface glycoprotein CD3 zeta chain Proteins 0.000 description 1
- 101000914514 Homo sapiens T-cell-specific surface glycoprotein CD28 Proteins 0.000 description 1
- 102000004877 Insulin Human genes 0.000 description 1
- 108090001061 Insulin Proteins 0.000 description 1
- 108010002350 Interleukin-2 Proteins 0.000 description 1
- 108020004684 Internal Ribosome Entry Sites Proteins 0.000 description 1
- 102000002698 KIR Receptors Human genes 0.000 description 1
- 108010043610 KIR Receptors Proteins 0.000 description 1
- 108010009254 Lysosomal-Associated Membrane Protein 1 Proteins 0.000 description 1
- 102100035133 Lysosome-associated membrane glycoprotein 1 Human genes 0.000 description 1
- ACFGRWJEQJVZTM-LEJBHHMKSA-L Magnesium L-ascorbic acid-2-phosphate Chemical compound [Mg+2].OC[C@H](O)[C@H]1OC(=O)C(OP([O-])([O-])=O)=C1O ACFGRWJEQJVZTM-LEJBHHMKSA-L 0.000 description 1
- 241001465754 Metazoa Species 0.000 description 1
- 208000034578 Multiple myelomas Diseases 0.000 description 1
- 229930182555 Penicillin Natural products 0.000 description 1
- JGSARLDLIJGVTE-MBNYWOFBSA-N Penicillin G Chemical compound N([C@H]1[C@H]2SC([C@@H](N2C1=O)C(O)=O)(C)C)C(=O)CC1=CC=CC=C1 JGSARLDLIJGVTE-MBNYWOFBSA-N 0.000 description 1
- 206010035226 Plasma cell myeloma Diseases 0.000 description 1
- 239000012979 RPMI medium Substances 0.000 description 1
- 108010008281 Recombinant Fusion Proteins Proteins 0.000 description 1
- 102000007056 Recombinant Fusion Proteins Human genes 0.000 description 1
- 239000006146 Roswell Park Memorial Institute medium Substances 0.000 description 1
- 102100037906 T-cell surface glycoprotein CD3 zeta chain Human genes 0.000 description 1
- 102100034922 T-cell surface glycoprotein CD8 alpha chain Human genes 0.000 description 1
- 102100027213 T-cell-specific surface glycoprotein CD28 Human genes 0.000 description 1
- 210000001744 T-lymphocyte Anatomy 0.000 description 1
- RYYWUUFWQRZTIU-UHFFFAOYSA-N Thiophosphoric acid Chemical class OP(O)(S)=O RYYWUUFWQRZTIU-UHFFFAOYSA-N 0.000 description 1
- 102000004338 Transferrin Human genes 0.000 description 1
- 108090000901 Transferrin Proteins 0.000 description 1
- 241000711975 Vesicular stomatitis virus Species 0.000 description 1
- 230000000735 allogeneic effect Effects 0.000 description 1
- 210000000270 basal cell Anatomy 0.000 description 1
- 230000029918 bioluminescence Effects 0.000 description 1
- 238000005415 bioluminescence Methods 0.000 description 1
- 238000013406 biomanufacturing process Methods 0.000 description 1
- 210000004369 blood Anatomy 0.000 description 1
- 210000001185 bone marrow Anatomy 0.000 description 1
- 230000030833 cell death Effects 0.000 description 1
- 230000011712 cell development Effects 0.000 description 1
- 230000032823 cell division Effects 0.000 description 1
- 238000002659 cell therapy Methods 0.000 description 1
- 230000003833 cell viability Effects 0.000 description 1
- 239000003153 chemical reaction reagent Substances 0.000 description 1
- 239000011248 coating agent Substances 0.000 description 1
- 238000000576 coating method Methods 0.000 description 1
- 238000011109 contamination Methods 0.000 description 1
- 231100000433 cytotoxic Toxicity 0.000 description 1
- 230000001472 cytotoxic effect Effects 0.000 description 1
- 230000034994 death Effects 0.000 description 1
- 231100000517 death Toxicity 0.000 description 1
- 230000003247 decreasing effect Effects 0.000 description 1
- 239000005547 deoxyribonucleotide Substances 0.000 description 1
- 125000002637 deoxyribonucleotide group Chemical group 0.000 description 1
- 238000009795 derivation Methods 0.000 description 1
- 201000010099 disease Diseases 0.000 description 1
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 1
- NAGJZTKCGNOGPW-UHFFFAOYSA-N dithiophosphoric acid Chemical class OP(O)(S)=S NAGJZTKCGNOGPW-UHFFFAOYSA-N 0.000 description 1
- 230000002708 enhancing effect Effects 0.000 description 1
- 230000001973 epigenetic effect Effects 0.000 description 1
- 239000012467 final product Substances 0.000 description 1
- 238000007667 floating Methods 0.000 description 1
- 239000012530 fluid Substances 0.000 description 1
- 230000030279 gene silencing Effects 0.000 description 1
- 238000010353 genetic engineering Methods 0.000 description 1
- PCHJSUWPFVWCPO-UHFFFAOYSA-N gold Chemical compound [Au] PCHJSUWPFVWCPO-UHFFFAOYSA-N 0.000 description 1
- 206010073071 hepatocellular carcinoma Diseases 0.000 description 1
- 102000056003 human IL15 Human genes 0.000 description 1
- 230000001771 impaired effect Effects 0.000 description 1
- 238000010348 incorporation Methods 0.000 description 1
- 238000003780 insertion Methods 0.000 description 1
- 230000037431 insertion Effects 0.000 description 1
- 229940125396 insulin Drugs 0.000 description 1
- 238000002955 isolation Methods 0.000 description 1
- 238000011031 large-scale manufacturing process Methods 0.000 description 1
- 208000014018 liver neoplasm Diseases 0.000 description 1
- 230000007774 longterm Effects 0.000 description 1
- 230000007246 mechanism Effects 0.000 description 1
- 230000001404 mediated effect Effects 0.000 description 1
- 239000013028 medium composition Substances 0.000 description 1
- 108020004999 messenger RNA Proteins 0.000 description 1
- 230000011987 methylation Effects 0.000 description 1
- 238000007069 methylation reaction Methods 0.000 description 1
- YACKEPLHDIMKIO-UHFFFAOYSA-N methylphosphonic acid Chemical class CP(O)(O)=O YACKEPLHDIMKIO-UHFFFAOYSA-N 0.000 description 1
- 238000013508 migration Methods 0.000 description 1
- 230000005012 migration Effects 0.000 description 1
- 238000002156 mixing Methods 0.000 description 1
- 238000012544 monitoring process Methods 0.000 description 1
- 230000007819 multidimensional cell growth Effects 0.000 description 1
- 208000025113 myeloid leukemia Diseases 0.000 description 1
- 238000010899 nucleation Methods 0.000 description 1
- 230000009437 off-target effect Effects 0.000 description 1
- 230000000771 oncological effect Effects 0.000 description 1
- 230000003287 optical effect Effects 0.000 description 1
- 238000005457 optimization Methods 0.000 description 1
- 230000002018 overexpression Effects 0.000 description 1
- 229940049954 penicillin Drugs 0.000 description 1
- 230000002093 peripheral effect Effects 0.000 description 1
- 150000008298 phosphoramidates Chemical class 0.000 description 1
- 239000013612 plasmid Substances 0.000 description 1
- 238000007747 plating Methods 0.000 description 1
- 108091033319 polynucleotide Proteins 0.000 description 1
- 102000040430 polynucleotide Human genes 0.000 description 1
- 239000002157 polynucleotide Substances 0.000 description 1
- 239000002243 precursor Substances 0.000 description 1
- 230000002265 prevention Effects 0.000 description 1
- 230000001737 promoting effect Effects 0.000 description 1
- 102000004169 proteins and genes Human genes 0.000 description 1
- 238000000746 purification Methods 0.000 description 1
- 238000011160 research Methods 0.000 description 1
- 230000004044 response Effects 0.000 description 1
- 230000000717 retained effect Effects 0.000 description 1
- 125000002652 ribonucleotide group Chemical group 0.000 description 1
- 150000003839 salts Chemical class 0.000 description 1
- HVAKSLUOHARFLM-UHFFFAOYSA-N selenium;sodium Chemical compound [Se][Na] HVAKSLUOHARFLM-UHFFFAOYSA-N 0.000 description 1
- 230000011664 signaling Effects 0.000 description 1
- 229910000030 sodium bicarbonate Inorganic materials 0.000 description 1
- 239000000243 solution Substances 0.000 description 1
- 230000003068 static effect Effects 0.000 description 1
- 238000003860 storage Methods 0.000 description 1
- 238000007920 subcutaneous administration Methods 0.000 description 1
- 239000007929 subcutaneous injection Substances 0.000 description 1
- 238000010254 subcutaneous injection Methods 0.000 description 1
- 239000013589 supplement Substances 0.000 description 1
- 230000009897 systematic effect Effects 0.000 description 1
- 210000001519 tissue Anatomy 0.000 description 1
- 238000013518 transcription Methods 0.000 description 1
- 230000035897 transcription Effects 0.000 description 1
- 239000012581 transferrin Substances 0.000 description 1
- 230000007704 transition Effects 0.000 description 1
- 238000011269 treatment regimen Methods 0.000 description 1
- 230000004614 tumor growth Effects 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P37/00—Drugs for immunological or allergic disorders
- A61P37/02—Immunomodulators
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K2239/00—Indexing codes associated with cellular immunotherapy of group A61K39/46
- A61K2239/31—Indexing codes associated with cellular immunotherapy of group A61K39/46 characterized by the route of administration
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K2239/00—Indexing codes associated with cellular immunotherapy of group A61K39/46
- A61K2239/38—Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the dose, timing or administration schedule
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/46—Cellular immunotherapy
- A61K39/461—Cellular immunotherapy characterised by the cell type used
- A61K39/4613—Natural-killer cells [NK or NK-T]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/46—Cellular immunotherapy
- A61K39/463—Cellular immunotherapy characterised by recombinant expression
- A61K39/4631—Chimeric Antigen Receptors [CAR]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/46—Cellular immunotherapy
- A61K39/464—Cellular immunotherapy characterised by the antigen targeted or presented
- A61K39/4643—Vertebrate antigens
- A61K39/4644—Cancer antigens
- A61K39/464402—Receptors, cell surface antigens or cell surface determinants
- A61K39/464411—Immunoglobulin superfamily
- A61K39/464412—CD19 or B4
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/475—Growth factors; Growth regulators
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/475—Growth factors; Growth regulators
- C07K14/51—Bone morphogenetic factor; Osteogenins; Osteogenic factor; Bone-inducing factor
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/705—Receptors; Cell surface antigens; Cell surface determinants
- C07K14/70503—Immunoglobulin superfamily
- C07K14/7051—T-cell receptor (TcR)-CD3 complex
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N5/00—Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
- C12N5/06—Animal cells or tissues; Human cells or tissues
- C12N5/0602—Vertebrate cells
- C12N5/0634—Cells from the blood or the immune system
- C12N5/0646—Natural killers cells [NK], NKT cells
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N5/00—Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
- C12N5/06—Animal cells or tissues; Human cells or tissues
- C12N5/0602—Vertebrate cells
- C12N5/0634—Cells from the blood or the immune system
- C12N5/0647—Haematopoietic stem cells; Uncommitted or multipotent progenitors
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/63—Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
- C12N15/79—Vectors or expression systems specially adapted for eukaryotic hosts
- C12N15/85—Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
- C12N15/86—Viral vectors
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/10—Growth factors
- C12N2501/125—Stem cell factor [SCF], c-kit ligand [KL]
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/10—Growth factors
- C12N2501/155—Bone morphogenic proteins [BMP]; Osteogenins; Osteogenic factor; Bone inducing factor
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/10—Growth factors
- C12N2501/165—Vascular endothelial growth factor [VEGF]
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/20—Cytokines; Chemokines
- C12N2501/23—Interleukins [IL]
- C12N2501/2303—Interleukin-3 (IL-3)
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/20—Cytokines; Chemokines
- C12N2501/23—Interleukins [IL]
- C12N2501/2307—Interleukin-7 (IL-7)
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/20—Cytokines; Chemokines
- C12N2501/23—Interleukins [IL]
- C12N2501/2315—Interleukin-15 (IL-15)
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/20—Cytokines; Chemokines
- C12N2501/26—Flt-3 ligand (CD135L, flk-2 ligand)
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/70—Enzymes
- C12N2501/72—Transferases [EC 2.]
- C12N2501/727—Kinases (EC 2.7.)
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2506/00—Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells
- C12N2506/02—Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from embryonic cells
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2506/00—Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells
- C12N2506/45—Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from artificially induced pluripotent stem cells
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2510/00—Genetically modified cells
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2513/00—3D culture
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2740/00—Reverse transcribing RNA viruses
- C12N2740/00011—Details
- C12N2740/10011—Retroviridae
- C12N2740/16011—Human Immunodeficiency Virus, HIV
- C12N2740/16041—Use of virus, viral particle or viral elements as a vector
- C12N2740/16043—Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
Definitions
- a major roadblock to current cell immunotherapies in cancer treatment is the high concentration of effector cells required per dose.
- Various strategies in cultivating hiPSC- derived NK cells involve using an excessive number of plates or flasks for expansion of the final product, which can be a highly inefficient, time-consuming, and expensive process that also increases overall susceptibility to cell culture contamination.
- Other methods of expanding NK cells involve the use of cancer feeder-cells, which results in the hiPSC-NK product being declassified as GMP-grade and quality.
- the methods described herein demonstrate that the entire differentiation and expansion of hiPSCs to functionally mature NK cells can take place entirely within an enclosed bioreactor system.
- the methods described herein streamline the differentiation process and ensure consistent reproducibility and reliability of the resultant NK cells.
- the methods described herein are performed under xeno-free and feeder-free conditions. Consequently, the final derived cells are GMP-compliant and can be used in, e.g., clinical trials.
- One small scale-bioreactor can potentially yield >10-50 x 10 7 total NK cells. This refinement of this hiPSC to NK cell differentiation process allows the opportunity to scale-up enough high-grade cells to meet the stringent requirements needed to proceed with human clinical trials.
- the methods described herein are efficient and cost-effective for generating phenotypically mature, functional, clinical-grade NK cells for use in developing cancer immunotherapies.
- NK cells were observed and specified in medium suspension almost 10 days faster when compared to 2D-controls (FIGs. 3 A-H).
- 3D-bioreactor-derived NK cells demonstrate advanced maturation and development.
- Flow cytometry analysis to quantitate expression levels of CD56, a marker for mature NK cells demonstrated that the 3D bioreactor-derived cells were over 93% positive at day 27 of differentiation compared to 58% at day 27 of differentiation for 2D-generated cultures (FIGs. 4A-F and 5).
- expression levels were measured for NK lineage-specific markers CD94, NKp44, NKG2D, Lampl, and CD 16.
- NK lineage markers for differentiation day 27 bioreactor-generated cells were comparable or significantly exceed expression levels from two independent 2D-differentiation rounds that were cultured almost twice as long (53 days and 55 days, respectively) (FIGs. 6A-O, and Table 1). Culturing hiPSCs in a 3D bioreactor using the methods and culture media described herein can generate highly pure and phenotypically mature cells that are similar to primary NK cells.
- FIG. 1 is a summary of a 3D-directed in vitro differentiation protocol for NK cells derived from human induced pluripotent stem cells (hiPSC).
- hiPSCs are scaled-up from a 2D-monolayer to a 3D-small scale bioreactor cell culture system with Y27632 supplementation to enhance iPSC viability and improve sphere formation.
- Hematopoietic induction of iPSCs to progenitor cells of an NK lineage restricted fate takes 12 days.
- the 3D-cultures are transferred to NK cell differentiation conditions. Two weeks thereafter, mature and functional NK cells can be continuously collected from the medium for a period of about 35 days.
- FIGs. 2A-C show 3D-adaptation of hiPSCs from 2D-monolayer culture utilizing a small-scale bioreactor.
- FIG. 2A is a low magnification (lOx) photomicrograph of hiPSCs in 2D-monolayer culture.
- FIG. 2B is a photograph of a small-scale 30ml bioreactor flask.
- FIG. 2C is a photomicrograph of hiPSC-spheres 20-hours after 2D-to-3D adaptation (lOx magnification).
- FIGs. 3 A-H is a comparison of the differentiation potential between 2D- monolayer versus 3D-bioreactor generated hiPSC-NK cells.
- FIGs. 3 A-D are images of adherent-2D derived cell-spheres and derivatives at indicated time points after the initiation of NK differentiation (lOx). Spheres slowly flatten over time with premature NK-like cells observed in cell medium at differentiation day 23.
- FIGs. 3A-B 4x magnification;
- FIGs. 3C- D lOx magnification.
- FIGs. 3E-H are images of 3D bioreactor generated cell-spheres and derivatives. Immature NK-like cells are observed two days after NK differentiation initiation on day 14.
- FIGs. 3E-F 20x magnification;
- FIGs. 3G-H lOx magnification.
- FIGs. 4A-F show characterization of hiPSC-NK cells.
- Flow cytometry quantification indicate a correlated temporal pattern of expression for the hematopoietic stem cell progenitor marker CD34 (FIG. 4A) and mature NK cell markers CD45 and CD56 (FIGs. 4B-F).
- FIG. 5 shows comparative CD56-positive marker expression between 2D- monolayer and 3D-bioreactor generated cells at specific time points during the differentiation timeline.
- FIGs. 6A-0 show NK lineage flow cytometry panel comparison of 3D-bioreactor and 2D-monolayer derived NK cells.
- Top row Flow cytometry panel for bioreactor derived NK cells at differentiation day 27.
- Second and third row Flow cytometry panel of two independently conducted 2D-monolayer trials in generating NK cells at differentiation day 53 and day 55, respectively.
- FIGs. 7A-J shows phenotypic comparison of hiPSC-NK vs. CBNK.
- FIGs. 8A-D show functional in vitro killing. In vitro killing using unmodified iPSC-NKs against Jurkat cells (FIG. 8A), HeLa cells (FIG. 8B), and K562 cells (FIG. 8C) as determined using Incucyte Base Analysis immune cell-killing software for live cell analysis.
- FIG. 8D shows in vitro killing comparing unmodified primary CBNK and iPSC-NKs against K562 cells (effectortarget ratio of 1 :2). Different concentration of target cancer cells was incubated with hiPSC-NK cells at effectortarget ratios of 1 : 1 and 4:1. Data represented as mean ⁇ SEM.
- FIGs. 9A-J show genetic modification of mature hiPSC-NK cells using lentivirus.
- hiPSC-NK cells can be transduced with lentiviral -based vectors to express a CD19-CAR construct with high affinity.
- primary NK cells demonstrate a persistent resistance to genetic engineering after lentiviral transduction.
- FIGs. 10 A- J show genetic modification of mature hiPSC-NK cells using retrovirus. Both mature hiPSC-NK and primary NK cells can be transduced with retroviralbased vectors to express a CD19-CAR construct with high affinity.
- FIGs. 11 A-C show comparative functional analysis between unmodified and CAR19 hiPSC-NK against tumor targets. In vitro killing against Raji cells (FIG. 11 A), NALM6 cells (FIG. 1 IB), and CCRF cells (FIG. 11C) as determined using Incucyte’s immune cell-killing software for live cell analysis. Different concentration of target cancer cells was incubated with hiPSC-NK cells or CAR19-hiPSC NK cells at effectortarget ratios of 2: 1. Data represented as mean ⁇ SEM.
- FIGs. 12A-C show genetic modification of iPSCs using lentivirus.
- FIG. 12A The selection and colony-picking of a single iPSC-colony from the surface of the cell culture plate.
- FIG. 12B After 2 days in differentiation medium, CD19-expression can be observed in the removed colonies under fluorescent imaging. A wide variation in CD19-expressing positive clones derived from selected colonies observed (top and bottom row, yellow arrows and arrowheads).
- FIG. 12C After 5 days in differentiation medium, the picked colonies expressing CD 19 are capable of proliferating and maintaining expression in vitro (yellow arrowheads).
- FIGs. 13A-B show NK-differentiation of lenti-CAR19 iPSCs using 3D bioreactor on day 12.
- FIG. 13A Flow cytometry quantification for CD34 and CD45 marker expression. The untransduced hiPSC cell line exhibited 15% CD34-positive expression (middle panel) compared to 12% expression in the CAR19 S001 modified hiPSCs (far right panel). Percent positive for CD45 was ⁇ 1% in both cell lines.
- FIG. 13B CD19-CAR expression detected in differentiating spheres in 3D-bioreactor.
- FIG. 14 shows schematics of viral CAR constructs.
- FIGs. 15A-B show that cryopreserved unmodified hiPSC-derived NK cells demonstrate strong killing efficacy in a subcutaneous K562 myelogenous leukemia mouse model.
- FIG. 15 A Frozen peripheral blood derived NK (PBNK) or frozen iPSC-derived NK cells were injected intravenously (4 x 10 6 /mouse) post-tumor engraftment. Optical bioluminescence imaging of tumor load between days 3 and 7 post-treatment.
- FIG. 15B Quantification of tumor intensity examined across all control and experimental cohorts four days after NK-infusion.
- FIGs. 16A-B show iPSC-derived NK cells can survive and persist in vivo for at least two weeks post-injection.
- FIG. 16A Infused NK cells transduced with retroviral constructs expressing luciferase alone or luciferase + soluble IL 15 then were injected intravenously into the tail vein (2 x 10 6 /mouse).
- FIG. 16B Persistence of genetically modified iPSC-NK cells were detectable two weeks in vivo. Transduction of IL15 enhanced persistence of injected iPSC-NK cells, and there were no discernible effects on mice from iPSC-NK infusion.
- FIGs. 17A-D show that NK cells can be generated from a human embryonic stem cell (hESC) source using a 3D-bioreactor platform system.
- FIGs. 17A-C High magnification (40x) photomicrographs of cell suspensions collected from the 3D-bioreactor demonstrate a phenotypic maturation of NK cells over developmental time.
- FIGs. 17D A purified population of NK cells were collected and maintained over time as indicated by flow cytometry quantification of mature NK cell markers CD45 and CD56.
- FIGs. 18A-F NK cells generated from a human embryonic stem cell (hESC) source using a 3D-bioreactor platform system show potent endogenous killing activity and can be genetically modified to enhance killing activity using a CAR construct in hepatocellular cancer lines.
- FIG. 18 A ESC-CAR NK in vitro killing displayed as percent growth against Huh7 cancer cell line over a 24-hour period.
- FIG. 18B Average Huh7 percent cancer growth at 24 hours.
- FIG. 18C ESC-CAR NK in vitro killing displayed as percent growth against HepG2 cancer cell line over a 24-hour period.
- FIG. 18D Average HepG2 percent cancer growth at 24 hours.
- FIG.18E ESC-CAR NK in vitro killing displayed as percent growth against Hep3B cancer cell line over a 24-hour period.
- FIG. 18F Average Hep3B percent cancer growth at 24 hours.
- NK cells Natural Killer cells
- PBNK peripheral blood
- NK cells isolated from cord blood exhibit higher cell viability after cryopreservation, however, due to the lower number of NK cells detected per cord blood unit, numerous expansion phases are required to obtain multiple units per dose [4],
- CBNKs display a more immature phenotype that is directly correlated to a reduction in cytotoxicity [4, 5]
- cytotoxicity loss can be partially resolved through cytokine support and feeder cell co-culture, these considerations demonstrate that PBNKs and CBNKs are not ideal for an “off-the-shelf’ allogeneic immunotherapy whereby the NK cells are harvested from unrelated donors, cryopreserved, and thawed.
- hESCs Human embryonic stem cells
- iPSCs induced pluripotent stem cells
- An infinite starting cell source is a tremendous advantage in scaling-up the production of a homogenous and high-quality NK cell product.
- GMP-grade iPSC-derived NK cells This allows for the development and expansion of GMP-grade iPSC-derived NK cells to be used in cancer immunotherapy clinical trials.
- Several research groups have published recent advances in methodological strategies to successfully differentiate pluripotent stem cells into the NK lineage that display comparable phenotype and function to primary NK cells [6-10], Some of those methods involve differentiation of NK cells by co-culturing with OP9 mouse stromal cells.
- PSC-derived NK cells are a promising alternative approach to cancer immunotherapy.
- pluripotent stem cells includes human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs). The methods described here are suitable for use with pluripotent stem cells.
- a three-dimensional (3D) bioreactor promotes growth of cells in three dimensions.
- a 3D bioreactor is distinguished from two-dimensional (2D) growth of cells on a flat surface, such as a cell culture dish. By promoting cell growth in three dimensions, the cells can grow into spheroids, or 3D cell colonies.
- Some 3D bioreactors have scaffolds to which adherent cells can attach.
- Other 3D bioreactors employ scaffold-free techniques.
- One example of a scaffold free 3D bioreactor is a 30 ml disposable magnetic stir bioreactor from ABLE Corporation (Japan) and Biott Corporation (Japan) (ABLE Cat No. BWV-S03 A), which uses an impeller with a magnet on each blade to provide low-shear agitation by laminar flow, which can encourage the formation and growth of spheroid cell clusters.
- An impeller or other mechanism for mixing the culture media provides multiple benefits, including enhanced mass transfer and increased cell surface area exposure to media, nutrients, and differentiation factors.
- the 3D bioreactor is an enclosed bioreactor.
- Pluripotent stem cells are cultured in a cell culture medium that maintains them in a pluripotent state as the cells grow from an initial aliquot to a 3D culture.
- Many commercially-available feeder-free, xenofree culture media are suitable for use as a basal medium for iPSC expansion.
- One example is Essential 8 medium (available from ThermoFisher Scientific).
- Others include mTeSR PlusTM (StemCell Technologies);
- NutriStem® XF medium (Biological Industries); Cellartis DEF-CS 500 (Takara); and StemFlexTM Medium (Fisher Scientific).
- Another suitable medium is described in Chen, G. et al. Chemically defined conditions for human iPSC derivation and culture. Nat Methods. 2011;8(5):424-429 (Methods, Human ES Cell Culture, E8 media composition).
- the media described therein includes DMEM/F12, L-ascorbic acid-2-phosphate magnesium (64 mg/1), sodium selenium (14 pg/1), FGF2 (100 pg/1), insulin (19.4 mg/1), NaHCO 3 (543 mg/1) and transferrin (10.7 mg/1), TGFpi(2 pg/1) or NODAL (100 pg/1). Osmolarity of all media was adjusted to 340 mOsm at pH 7.4.
- concentrations need not be precisely as described in Chen, G. et al.
- the pluripotent stem cell expansion medium can also include vitronectin, which is a glycoprotein that provides a surface coating to promote cell attachment for use in feeder- free culture of pluripotent stem cells.
- the pluripotent stem cell expansion medium can also include a rho-kinase (ROCK) inhibitor.
- a ROCK inhibitor is only used when hiPSCs or hESCs need to be enzymatically passaged to help mitigate cell death.
- Y-27632 is a compound having the following structure:
- Y-27632 is sometimes available as a salt e.g., ⁇ 2 HC1).
- the cell density increases and the cells form 3D-PSC spheres after bioreactor adaptation.
- Expression of OCT- 4 a marker of pluripotency, of the 3D-PSCs can be quantified by flow cytometry.
- OCT-4 a marker of pluripotency
- the PSCs are typically cultured to a cell density of at least about 750,000 cells/mL. In some instances, the PSCs are cultured to a cell density from about 750,000 cells/mL to about 1,250,000 cells/mL. For the 30 mL bioreactor used in the Examples, the iPSCs were cultured to approximately 25 million cells to 35 million cells.
- the cells can be mechanically dissociated during cell culture (e.g., with a pipette) in order to separate cell clumps or aggregates into smaller clumps, smaller aggregates, or single suspended cells.
- HSCs hematopoietic stem cells
- the cells are then cultured to differentiate the 3D PSCs to hematopoietic stem cells (HSCs).
- HSCs hematopoietic stem cells
- the hematopoietic stem cell culture medium includes stem cell factor (SCF), bone morphogenetic protein 4 (BMP4) (or an agonist of BMP4), and vascular endothelial growth factor (VEGF).
- SCF stem cell factor
- BMP4 bone morphogenetic protein 4
- VEGF vascular endothelial growth factor
- the hematopoietic stem cell culture medium typically also includes a basal culture medium that promotes hematopoietic differentiation.
- One suitable basal culture medium is albumin polyvinylalcohol essential lipids (APEL). Suitable APEL culture media are described in US Patent No.
- the cells express CD34+, a marker of hematopoietic stem cells (HSCs).
- HSCs hematopoietic stem cells
- the cells are cultured for about 6 days to about 12 days. After about day 12, the cells begin to transition to a more mature (differentiated) cell type (e.g., natural killer cells). Once the cells are differentiated to HSCs, the cells can be differentiated to natural killer cells.
- BMP4 agonist is SB 4, which is a compound having the following structure:
- the cells are then cultured to differentiate them from hematopoietic stem cells (HSCs) to a differentiated cell type, such as natural killer cells.
- HSCs hematopoietic stem cells
- One example of a cell culture medium includes: i) Dulbecco's Modified Eagle Medium with Glutamax; ii) Dulbecco's Modified Eagle Medium/Nutrient Mixture F-12 with Glutamax containing Human AB serum; iii) ethanolamine; iv) 2-mercaptoethanol (2-BME); v) sodium selenite; and vi) ascorbic acid with interleukin 3 (IL3), interleukin 7 (IL7), interleukin (IL 15), FMS-like tyrosine kinase 3 ligand (Flt3L), stem cell factor (SCF).
- IL3 interleukin 3
- IL7 interleukin 7
- IL 15 interleukin
- FMS-like tyrosine kinase 3 ligand FMS-like tyrosine kinase 3 ligand
- SCF stem cell factor
- Human AB serum refers to Type AB serum typically derived from male donors. Type AB donors lack antibodies against A and B-blood type antigens, therefore more commonly used to mitigate immunoreactivity.
- FMS-like tyrosine kinase 3 ligand can enhance IL15 signaling, which can be important for the NK lineage.
- IL15 is a cytokine involved in the specific maintenance and proliferation of NK cells, and thus, Flt3L may enhance proliferation and/or NK specification as a result.
- NK cells As cells mature and adapt an NK-lineage identity, the NK cells are released into the cell medium, from which they can be collected for use in downstream applications. Cells can be cultured indefinitely in this cell culture medium as NK cells are produced. Lentiviral transduction
- the lentivirus is concentrated with Lenti-X Concentrator (Takara Bio, Cat. Nos. 631231 & 631232).
- RetroNectin is a 63 kD fragment of recombinant human fibronectin fragment (also referred to as rFN-CH-296).
- Lentiviral transduction provides advantages relative to retroviral transduction.
- Lentiviruses LVs
- retroviruses can only infect actively dividing, mitotically active cells. Accordingly, transduction with lentivirus is, in theory, more efficient as it has the potential to incorporate into all NK cells as opposed to only proliferating ones.
- NK cells are resistant to LV transduction, which hampers their development as an immunotherapy.
- VSV-G Vesicular Stomatitis Virus type-G
- CAR chimeric antigen receptor
- nucleic acid As used herein, the terms “nucleic acid,” “nucleotide,” and “polynucleotide” shall be given their ordinary meanings and shall include deoxyribonucleotides, deoxyribonucleic acids, ribonucleotides, and ribonucleic acids, and polymeric forms thereof, and includes either single- or double-stranded forms. Nucleic acids include naturally occurring nucleic acids, such as deoxyribonucleic acid (“DNA”) and ribonucleic acid (“RNA”) as well as nucleic acid analogs.
- DNA deoxyribonucleic acid
- RNA ribonucleic acid
- Nucleic acid analogs include those which include non-naturally occurring bases, nucleotides that engage in linkages with other nucleotides other than the naturally occurring phosphodiester bond or which include bases attached through linkages other than phosphodiester bonds.
- nucleic acid analogs include, for example and without limitation, phosphorothioates, phosphorodithioates, phosphorotriesters, phosphoramidates, boranophosphates, methylphosphonates, chiral-methyl phosphonates, 2-O-methyl ribonucleotides, peptide-nucleic acids (PNAs), locked-nucleic acids (LNAs), and the like.
- operably linked for example in the context of a regulatory nucleic acid sequence being “operably linked” to a heterologous nucleic acid sequence, shall be given its ordinary meaning and shall mean that the regulatory nucleic acid sequence is placed into a functional relationship with the heterologous nucleic acid sequence.
- “operably linked to” refers to a functional linkage between a nucleic acid sequence containing an internal ribosome entry site and a heterologous coding sequence initiation in the middle of an mRNA sequence resulting in translation of the heterologous coding sequence.
- vector shall be given its ordinary meaning and shall refer to a vehicle by which a DNA or RNA sequence (e.g., a foreign gene) can be introduced into a genetically engineered cell, so as to transform the genetically engineered cell and promote expression (e.g., transcription and/or translation) of the introduced sequence.
- a DNA or RNA sequence e.g., a foreign gene
- Embodiments described herein related to transduction of cells to express a chimeric antigen receptor can be transduced by a vector, such as a lentiviral vector, to express a chimeric antigen receptor.
- a lentiviral vector can include a nucleic acid sequence encoding a chimeric antigen receptor.
- Chimeric antigen receptors include an extracellular antigen recognition domain, a transmembrane domain, and an intracellular activation domain.
- the extracellular antigen recognition domain can be, for example, a single-chain Fragment variant (scFv) derived from an antibody. In embodiments described herein, the extracellular antigen recognition domain binds CD 19.
- scFv single-chain Fragment variant
- the transmembrane domain can be, for example, a CD3 transmembrane domain, a CD8 transmembrane domain, or a CD28 transmembrane domain.
- the intracellular activation domain can be, for example, a CD3 ⁇ domain (sometimes written as CD3-zeta, CD3Z, or CD3z).
- the chimeric antigen receptor can include a co-stimulatory domain, such as, for example, a 4- IBB domain.
- hiPSC cell lines were obtained from Al stem that were derived from umbilical cord blood. Human embryonic stem cells (hESCs) were acquired from ESI BIO (Alameda, California, USA). hiPSC culture
- hiPSCs and hESCs were cultured and maintained in Essential 8 medium (ThermoFisher Scientific, Catalog# A2858501).
- the supplementation of the ROCK Inhibitor Y27632 (Sigma, Cat# SCM075) was added to hiPSC and hESC cultures at lOpM during cell passaging only and removed from the medium within 24 hours of plating.
- STEMdiff APEL 2 albumin, polyvinyl alcohol, essential lipids; Stemcell Technologies, Cat# 05275
- basal cell culture medium supplemented with Stem Cell Factor (R&D Systems, SCF, 40ng/ml, Cat# 255-SC/CF-050), Bone Morphogenetic Protein 4 (R&D Systems, BMP4, 20ng/ml, Cat# 314-BP-050) and Vascular Endothelial Growth Factor (R&D Systems, VEGF, 20ng/ml, Cat# 293-VE-050).
- Stem Cell Factor R&D Systems, SCF, 40ng/ml, Cat# 255-SC/CF-050
- Bone Morphogenetic Protein 4 R&D Systems, BMP4, 20ng/ml, Cat# 314-BP-050
- Vascular Endothelial Growth Factor R&D Systems, VEGF, 20ng/ml, Cat# 293-VE-050
- Basal medium is composed of a mixture of 56.6% DMEM + Glutamax (Life Technologies, Cat# 10566-016) and 28.3% F-12 + Glutamax (Life Technologies, Cat# 31765035) supplemented with 15% human heat-inactivated type-AB serum (Valley Biomedical, Cat# HP1022 HI), 50pM Ethanolamine (MP Biomedicals, Cat# 194658), 25pM 2-Mercaptoethanol (Sigma, Cat# M6250), 5ng/ml sodium selenite (Sigma, Cat# S5261), 20mg/ml ascorbic acid (Sigma, Cat# A-5960), and 1% penicillin-streptomycin (Gibco, Cat# 15140-148).
- Cytokine addition into the medium consisted of 5ng/ml IL-3 (Peprotech, Cat# 200-03), 20ng/ml IL-7 (Peprotech, Cat# 200-07), lOng/ml IL-15 (Peprotech, Cat# 200-15), lOng/ml Flt3 ligand (Peprotech, Flt3L, Cat# 300-19), and 20ng/ml SCF.
- 5ng/ml IL-3 Peprotech, Cat# 200-03
- 20ng/ml IL-7 Peprotech, Cat# 200-07
- lOng/ml IL-15 Peprotech, Cat# 200-15
- lOng/ml Flt3 ligand Peprotech, Flt3L, Cat# 300-19
- SCF 20ng/ml SCF
- Undifferentiated feeder-free hiPSCs and hESCs are cultured on vitronectin coated substrates in Essential 8 medium. When hiPSCs and hESCs cultures are >70% confluent, cells can be enzymatically passaged using either TrypLE or Accutase and re-seeded in a split ratio between 1 : 10 and 1 : 15 with Y27632 supplementation at a final concentration of lOpM. hiPSCs and hESCs can be continually maintained, expanded, or cryopreserved under these outlined conditions. Prior to initiating 2D-monolayer to 3D-bioreactor hiPSC adaptation, hiPSCs and hESCs need to be expanded until sufficient cells needed have been obtained.
- hiPSCs and hESCs are transferred as a starting 2D-monolayer into small-scale bioreactors to initiate 3D-suspension culture.
- 2D-hiPSCs and hESCs expansion has yielded a cell concentration of more than about 30 x 10 6 live cells
- the undifferentiated hiPSCs and hESCs can be dissociated with TrypLE or Accutase.
- Cell contents should be filtered through a 70pM cell strainer to remove doublets.
- Optimal seeding density into bioreactors should be approximately 1 x 10 6 cells/ml in Essential 8 medium with Y27632 supplementation at a final concentration of 10 pM.
- the rotational speed of the bioreactor at all phases of the 3D-differentiation process ranges from 60-80 RPM. Eighteen to twenty-four hours after adaptation, spheres should be examined carefully under the microscope and assessed for viability and morphology. Hematopoietic lineage induction
- the induction of pluripotent stem cells (hiPSCs and hESCs) towards a hematopoietic fate can be initiated after successful 3D- adaptation performed from the previous day.
- the complete removal of Essential 8 medium from the suspension culture is critical and can be executed by transferring the entire volume from the bioreactor to a 50ml conical tube. Leave cells undisturbed for 10-12 minutes to let the spheres pellet to the bottom. Remove the cell supernatant leaving the pellet undisturbed and replace with Hematopoietic induction medium described previously.
- Stemdiff APEL 2 medium supplemented with SCF at a concentration of 40ng/ml in conjunction with BMP4 and VEGF at a concentration of 20ng/ml each.
- Medium change and preparation can be conducted as described during hematopoietic induction stage between differentiation days 1 to 12.
- the expression of the hematopoietic stem cell progenitor marker CD34 and mature NK marker CD45 can be quantified at Day 6 and Day 12 to analyze the progression of NK cell development over time.
- the 3D-sphere aggregates can be further differentiated into the NK cell lineage by replacing the cultures with the above- mentioned NK cell differentiation medium.
- the NK cell differentiation base medium is composed of 56.6% DMEM + Glutamax and 28.3% F-12 + Glutamax supplemented with 15% human heat-inactivated type- AB serum, 50pM Ethanolamine, 25 pM 2- Mercaptoethanol, 5ng/ml sodium selenite, 20mg/ml ascorbic acid, and 1% penicillinstreptomycin.
- Cytokine addition into the medium consisted of 5ng/ml IL-3 (Peprotech, Cat# 200-03), 20ng/ml IL-7, lOng/ml IL- 15, lOng/ml Flt3 ligand, and 20ng/ml SCF. From differentiation day 12 and onwards, only NK differentiation medium is required. Medium in the 3D-bioreactor cultures should be replaced daily or when needed.
- pluripotent stem cell-derived NK cells delaminate from the differentiating 3D-sphere aggregates and are released into the cell culture medium suspension after detachment. Mature appearing NK cells can be observed floating in the cell medium after 1 week of NK cell differentiation.
- hiPSC derived NK cells can be collected directly from the medium and phenotyped for surface antigens that include CD45, CD56, CD 16, KIR, CD94, NKp44, NKG2D, and Lampl using flow cytometry. Functional analyses of hiPSC- derived NK cells can be assessed by in vitro killing assays against numerous cell cancer lines at various effector to target (E:T) ratios. NK cells developed using this 3D-bioreactor platform demonstrate a mature NK cell lineage profile and potent cell cytotoxicity.
- the methods outlined indicate an efficient and streamlined approach in deriving mature and functional NK cells from a pluripotent stem cells source that can be used as an infinite cell source for potential cancer cell therapies.
- the cancer target cells are labeled with the Incucyte Nuclight Red lentivirus, which enables the counting of viable cancer cells over time.
- Puromycin is added to the cancer cell cultures on day 6 at a concentration of 0.5ug/ml to initiate the selection process. Monitor transduction efficiency daily and supplement with puromycin into the cell culture medium with every media change. By day 10-14, a high percentage of the cancer target cells should be Nuclight red.
- Both effector and target cells are cultured in RPMI medium supplemented with 10% FBS for cytotoxicity analysis.
- Real-time NK cell cytotoxicity was monitored and quantified using Incucyte Base Analysis Software.
- Retronectin and wash wells with complete RPMI media Add 2ml of concentrated virus to Retronectin-coated wells. Spin plate at 2000 g for at least 1 hr at room temperature. Prepare NK MACs media with 500IU/ml IL-2 and 140IU/ml IL-15. Add 2.5E5 cells/ml (1ml per 24 well). Culture undisturbed for a minimum of 2 days before subsequent assays. Monitor CAR expression using flow cytometry and use transduced iNKs for subsequent killing assays.
- Subcutaneous injections of K562 cancer cells were prepared at 2 x 10 5 cells/mouse. K562 cancer cells were allowed to engraft for 3 days before systematic infusion of hiPSC-NK cells. The cryopreserved hiPSC-NK cells were thawed and prepared for infusion on the same day. Sample preparation for both hiPSC-NK and PBNK were identical as 4 x 10 6 NK cells/mouse were intravenously injected. Intraperitoneal (IP) injections of recombinant human IL-15 at 30 ng/mouse was administered daily for days 1-7 after NK cell infusion.
- IP intraperitoneal
- FIG. 1 is a summary of a 3D-directed in vitro differentiation protocol for NK cells derived from human induced pluripotent stem cells (hiPSC)
- hiPSC human induced pluripotent stem cells
- the induction of hiPSCs towards a hematopoietic fate can be initiated after successful 3D-hiPSC adaptation, which requires approximately 12 days, after which time 3D- cultures are transferred to NK cell differentiation conditions.
- Two weeks thereafter, mature and functional NK cells can be continuously collected from the medium for a period of about 35 days.
- hiPSCs were scaled-up from a 2D-monolayer to a 3D-small scale bioreactor cell culture system with Y27632 supplementation to enhance iPSC viability and improve sphere formation.
- hiPSCs were grown as 2D-monolayer cultures to a starting concentration of 30 x 10 6 live cells. These undifferentiated hiPSCs were dissociated with TrypLE then filtered through a 70pM cell strainer to achieve single cell suspension. The 3D-adaptation of hiPSCs from 2D-monolayer culture was completed utilizing a small-scale bioreactor.
- FIG. 2A is a low magnification (lOx) photomicrograph of hiPSCs in 2D-monolayer culture.
- FIG. 2B is a photograph of a small-scale 30 ml bioreactor flask.
- FIG. 2C is a photomicrograph of hiPSC- spheres 20-hours after 2D-to-3D adaptation.
- FIGs. 3A-D are images of adherent-2D derived cell-spheres and derivatives at indicated time points after the initiation of NK differentiation (lOx). Spheres slowly flatten over time with premature NK-like cells observed in cell medium at differentiation day 23.
- FIGs. 3E-H are images of 3D bioreactor generated cell-spheres and derivatives. Immature NK-like cells are observed two days after NK differentiation initiation on day 14.
- 3D-bioreactor-derived NK cells demonstrate advanced maturation and development.
- Flow cytometry quantification indicates a correlated temporal pattern of expression for the hematopoietic stem cell progenitor marker CD34 (FIG. 4A) and NK cell markers CD45 and CD56 (FIGs. 4B-F).
- CD34 hematopoietic stem cell progenitor marker
- FIGs. 4B-F NK cell markers
- Expression levels of CD56 a marker for mature NK cells, demonstrated that the 3D bioreactor-derived cells were over 93% positive at day 27 of differentiation compared to 58% at day 27 of differentiation for 2D-generated cultures (FIGs. 5A-F and 5).
- Comparison of expression levels of CD56 over time reveals that bioreactor- derived cells express consistently higher levels of CD56 across all timepoints tested as compared to similarly staged 2D samples (FIG. 5).
- NK lineage-specific markers CD94, NKp44, NKG2D, Lampl, and CD 16.
- Expression levels of these NK lineage markers for differentiation day 27 bioreactor-generated cells were comparable or significantly exceed expression levels from two independent 2D- differentiation rounds that were cultured almost twice as long (53 days and 55 days, respectively) (FIG 6, and Table 1).
- 3D-bioreactor-derived hiPSC-NK cells were compared to primary cord blood-derived NK (CBNK) cells (FIGs. 7A-J).
- hiPSC-NK cells expressed similar levels of mature NK markers CD56, CD 16, CD94, NKG2D, NKp44 and LAMP1 as compared to primary CBNK cells.
- CBNK primary cord blood-derived NK
- NK cells While it is important that NK cells express appropriate maturation markers, the gold standard for validating the phenotype of these cells is by assessing the functional killing capacity of the final NK product.
- Functional analyses of hiP SC -derived NK cells were assessed by in vitro killing assays against Jurkat (FIG. 8A), HeLa (FIG. 8B), and K562 (FIG. 8C) cancer cells as determined using using Incucyte Base Analysis immune killing software for live cell analysis. Different concentrations of target cancer cells were incubated with hiPSC-NK cells at effectortarget (E:T) ratios of 1 : 1 and 4: 1.
- E:T effectortarget
- NK cells developed using this 3D-bioreactor platform demonstrate a mature NK cell lineage profile and potent cell cytotoxicity.
- Virus transduction of pre-differentiated hiPSC-NK expanded under feeder-free conditions [0094]
- One method for incorporating genetic modifications into iPSC-NKs is via transduction of iPSCs followed by selection of single cell clones to create stable iPSC lines.
- this method is not ideal for high throughput screening purposes as the timeline for stable iPSC generation and subsequent NK differentiation is relatively long.
- the transduction protocol described herein was used to incorporate both lentiviral and retroviral constructs into pre-differentiated hiPSC-NK cells.
- NK cells Genetic modification of NK cells can be performed to improve their function, target specific antigens, and/or enhance persistence in vivo.
- One application in the development of NK therapeutics is the incorporation of different chimeric antigen receptor (CAR) constructs. Introducing CAR constructs into NK cells has been hindered by the relative difficulty of viral modification of NK cells in comparison with T cells.
- CAR chimeric antigen receptor
- Use of retroviral vectors for NK transduction has become more common. As retroviruses specifically infect proliferating cells, substantial efforts have been made to enhance NK proliferation as a means of enhancing transduction efficiency.
- lentiviral vectors can be advantageous because they reduce the potential for genomic insertion and potential off-target effects.
- transduction of NK cells with lentiviral vectors has been significantly more challenging than transduction with retroviral vectors.
- hiPSC-NK cells as well as primary adult peripheral blood- derived NK (PBNK) cells were transduced with either lentivirus (FIGs. 9A-J) or retrovirus (FIGs. 10A-J) using the protocol described herein.
- hiPSC-NK cells can be transduced with both retroviral and lentiviral -based vectors to express a CD19-CAR construct with high affinity.
- primary PBNK cells demonstrate a persistent resistance to lentiviral transduction. Schematics of the constructs are depicted in FIG. 14. Functional in vitro killing capacity of pre-differentiated hiPSC-NK cells transduced with CAR19
- 3D-bioreactor derived hiPSC-NKs were used to transduce a retroviral construct expressing both CAR19 as well as an IL- 15 overexpression cassette using the transduction method described herein. Resulting CAR19 expression was approximately 47%.
- Nontransduced (NT) or CAR19 hiPSC-NKs were subjected to an in vitro killing assay against CD19+ Raji cells (FIG. 11 A), CD19+ NALM6 cells (FIG. 1 IB), and CD19-CCRF cells (FIG. 11C) as determined using Incucyte’s immune cell-killing software for live cell analysis.
- CAR19-transduced hiPSC-NK cells showed enhanced killing efficiency against cancer cell lines expressing the CD 19 target.
- a preferred method of incorporating genetic modifications into iPSC-NKs is via transduction of iPSCs followed by selection of single cell clones to create stable iPSC lines. Stably introducing CAR constructs at the iPSC-stage, allows for the persistence of this genetic modification in all differentiated progenitors, resulting in an ultimate CAR transduction efficiency of 100%. Importantly, some genetic modifications induced during the iPSC-stage may result in impaired differentiation into the desired cell type.
- lentiviral transduction was performed on iPSC colonies, which were subsequently expanded as undifferentiated iPSCs for subsequent differentiation within the enclosed bioreactor system (FIGs. 12A-C). Single colonies from iPSC cultures were selected and replated onto new vitronectin substrates in the presence of concentrated lentivirus expressing a CAR19 construct (FIG. 12 A). After 2 days in differentiation medium, CAR19-expression can be observed in the removed colonies under fluorescent imaging.
- both nontransduced (NT) and CAR19 hiPSC lines were induced to differentiate into NK cells using the 3D bioreactor protocol.
- This approach allowed for the continued monitoring of the response of genetically modified hiPSC lines to the previously established 3D bioreactor differentiation protocol.
- separate bioreactors were used to differentiate both NT and CAR19 hiPSC lines.
- hematopoietic differentiation as determined by CD34 and CD45 expression were unchanged between NT and CAR19 hiPSC lines (FIG. 13 A).
- NT hiPSC cell line exhibited 15% CD34-positive expression (FIG.
- the methods described herein are performed under xeno-free and feeder-free conditions. Consequently, the final derived cells are GMP-compliant and can be used in, e.g., clinical trials.
- One small-scale (30 ml) bioreactor can potentially yield >10 x 10 7 to 50 x 10 7 total NK cells. This refinement of this hiPSC to NK cell differentiation process allows the opportunity to scale-up enough highgrade cells to meet the stringent requirements needed to proceed with human clinical trials.
- the methods described herein are efficient and cost-effective for generating phenotypically mature, functional, clinical-grade NK cells for use in developing cancer immunotherapies. Phenotypic characterization and killing efficacy of 3D-bioreactor derived embryonic stem cell (ESC)-NK cells
- Embryonic stem cells also generated NK cells at a similar relative efficiency to iPSCs following the differentiation protocol in the 3D-bioreactor platform system as previously described.
- a purified population of NK cells that express mature NK lineage markers were collected in suspension from the bioreactor medium (FIGs. 17A-D).
- These ESC-NK cells demonstrate robust in vitro killing against HUH7, HEPG2, and HEP3B cancer cell lines compared to the PBNK-control (FIGs. 18A-F).
- ESC-NK cells genetically modified to express a CAR (ESC-NK CAR) demonstrated a greater reduction in HUH7, HEPG2, and HEP3B cancer cells compared to non-transformed ESC-NK cells (ESC-NK NT).
- hiPSC-NK cells were transduced with retroviral constructs expressing luciferase alone or luciferase + secreted IL15, using methods described herein.
- Transduced hiPSC-NK cells were intravenously infused via tail vein injection (2 x 10 6 /mouse) then tracked over time using bioluminescent imaging.
- These genetically modified hiPSC-NK cells persisted in vivo for up to two weeks (FIGs. 16A-B).
- Those hiPSC-NK cells transduced with IL- 15 demonstrated enhanced persistence in vivo. Importantly, there were no discernible effects on mice resulting from the hiPSC-NK infusion.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Immunology (AREA)
- General Health & Medical Sciences (AREA)
- Organic Chemistry (AREA)
- Engineering & Computer Science (AREA)
- Cell Biology (AREA)
- Medicinal Chemistry (AREA)
- Public Health (AREA)
- Animal Behavior & Ethology (AREA)
- Veterinary Medicine (AREA)
- Zoology (AREA)
- Genetics & Genomics (AREA)
- Biomedical Technology (AREA)
- Microbiology (AREA)
- Epidemiology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Pharmacology & Pharmacy (AREA)
- Biotechnology (AREA)
- Mycology (AREA)
- Wood Science & Technology (AREA)
- Biochemistry (AREA)
- Hematology (AREA)
- Molecular Biology (AREA)
- General Engineering & Computer Science (AREA)
- Toxicology (AREA)
- Gastroenterology & Hepatology (AREA)
- Biophysics (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Oncology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Developmental Biology & Embryology (AREA)
- Orthopedic Medicine & Surgery (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
Abstract
Cell culture methods and media for expansion of pluripotent stem cells, such as induced pluripotent stem cells (iPSCs) and embryonic stem cells (ESCs), to a three- dimensional culture and differentiation to hematopoietic stem cells are described. The hematopoietic stem cells can be further differentiated to natural killer cells or macrophages. The natural killer cells can be further transformed to express a chimeric antigen receptor (CAR).
Description
Three-Dimensional Culturing of Pluripotent Stem Cells to Produce Hematopoietic Stem Cells
RELATED APPLICATION
[0001] This application claims the benefit of U.S. Provisional Application No. 63/232,911, filed on August 13, 2021. The entire teachings of the above application are incorporated herein by reference.
BACKGROUND
[0002] A major roadblock to current cell immunotherapies in cancer treatment is the high concentration of effector cells required per dose. Various strategies in cultivating hiPSC- derived NK cells involve using an excessive number of plates or flasks for expansion of the final product, which can be a highly inefficient, time-consuming, and expensive process that also increases overall susceptibility to cell culture contamination. Other methods of expanding NK cells involve the use of cancer feeder-cells, which results in the hiPSC-NK product being declassified as GMP-grade and quality.
SUMMARY
[0003] The methods described herein demonstrate that the entire differentiation and expansion of hiPSCs to functionally mature NK cells can take place entirely within an enclosed bioreactor system. The methods described herein streamline the differentiation process and ensure consistent reproducibility and reliability of the resultant NK cells.
[0004] In comparison to other known methods, the methods described herein are performed under xeno-free and feeder-free conditions. Consequently, the final derived cells are GMP-compliant and can be used in, e.g., clinical trials. One small scale-bioreactor can potentially yield >10-50 x 107 total NK cells. This refinement of this hiPSC to NK cell differentiation process allows the opportunity to scale-up enough high-grade cells to meet the stringent requirements needed to proceed with human clinical trials. Additionally, the methods described herein are efficient and cost-effective for generating phenotypically mature, functional, clinical-grade NK cells for use in developing cancer immunotherapies.
[0005] NK cells were observed and specified in medium suspension almost 10 days faster when compared to 2D-controls (FIGs. 3 A-H). In addition, 3D-bioreactor-derived NK cells demonstrate advanced maturation and development. Flow cytometry analysis to quantitate
expression levels of CD56, a marker for mature NK cells, demonstrated that the 3D bioreactor-derived cells were over 93% positive at day 27 of differentiation compared to 58% at day 27 of differentiation for 2D-generated cultures (FIGs. 4A-F and 5). To further assess the robustness and timing of cellular differentiation between the 3D-bioreactor and 2D- monolayer platforms, expression levels were measured for NK lineage-specific markers CD94, NKp44, NKG2D, Lampl, and CD 16. Expression levels of these NK lineage markers for differentiation day 27 bioreactor-generated cells were comparable or significantly exceed expression levels from two independent 2D-differentiation rounds that were cultured almost twice as long (53 days and 55 days, respectively) (FIGs. 6A-O, and Table 1). Culturing hiPSCs in a 3D bioreactor using the methods and culture media described herein can generate highly pure and phenotypically mature cells that are similar to primary NK cells.
BRIEF DESCRIPTION OF THE DRAWINGS
[0006] The foregoing will be apparent from the following more particular description of example embodiments, as illustrated in the accompanying drawings in which like reference characters refer to the same parts throughout the different views. The drawings are not necessarily to scale, emphasis instead being placed upon illustrating embodiments.
[0007] FIG. 1 is a summary of a 3D-directed in vitro differentiation protocol for NK cells derived from human induced pluripotent stem cells (hiPSC). Briefly, hiPSCs are scaled-up from a 2D-monolayer to a 3D-small scale bioreactor cell culture system with Y27632 supplementation to enhance iPSC viability and improve sphere formation. Hematopoietic induction of iPSCs to progenitor cells of an NK lineage restricted fate takes 12 days. Then, the 3D-cultures are transferred to NK cell differentiation conditions. Two weeks thereafter, mature and functional NK cells can be continuously collected from the medium for a period of about 35 days.
[0008] FIGs. 2A-C show 3D-adaptation of hiPSCs from 2D-monolayer culture utilizing a small-scale bioreactor. FIG. 2A is a low magnification (lOx) photomicrograph of hiPSCs in 2D-monolayer culture. FIG. 2B is a photograph of a small-scale 30ml bioreactor flask. FIG. 2C is a photomicrograph of hiPSC-spheres 20-hours after 2D-to-3D adaptation (lOx magnification).
[0009] FIGs. 3 A-H is a comparison of the differentiation potential between 2D- monolayer versus 3D-bioreactor generated hiPSC-NK cells. FIGs. 3 A-D are images of adherent-2D derived cell-spheres and derivatives at indicated time points after the initiation
of NK differentiation (lOx). Spheres slowly flatten over time with premature NK-like cells observed in cell medium at differentiation day 23. FIGs. 3A-B: 4x magnification; FIGs. 3C- D: lOx magnification. FIGs. 3E-H are images of 3D bioreactor generated cell-spheres and derivatives. Immature NK-like cells are observed two days after NK differentiation initiation on day 14. FIGs. 3E-F: 20x magnification; FIGs. 3G-H: lOx magnification.
[0010] FIGs. 4A-F show characterization of hiPSC-NK cells. Flow cytometry quantification indicate a correlated temporal pattern of expression for the hematopoietic stem cell progenitor marker CD34 (FIG. 4A) and mature NK cell markers CD45 and CD56 (FIGs. 4B-F).
[0011] FIG. 5 shows comparative CD56-positive marker expression between 2D- monolayer and 3D-bioreactor generated cells at specific time points during the differentiation timeline.
[0012] FIGs. 6A-0 show NK lineage flow cytometry panel comparison of 3D-bioreactor and 2D-monolayer derived NK cells. Mature NK cell expression markers assessed: CD56, CD94, NKp44, NKG2D, Lampl, and CD 16. Top row: Flow cytometry panel for bioreactor derived NK cells at differentiation day 27. Second and third row: Flow cytometry panel of two independently conducted 2D-monolayer trials in generating NK cells at differentiation day 53 and day 55, respectively.
[0013] FIGs. 7A-J shows phenotypic comparison of hiPSC-NK vs. CBNK.
[0014] FIGs. 8A-D show functional in vitro killing. In vitro killing using unmodified iPSC-NKs against Jurkat cells (FIG. 8A), HeLa cells (FIG. 8B), and K562 cells (FIG. 8C) as determined using Incucyte Base Analysis immune cell-killing software for live cell analysis. FIG. 8D shows in vitro killing comparing unmodified primary CBNK and iPSC-NKs against K562 cells (effectortarget ratio of 1 :2). Different concentration of target cancer cells was incubated with hiPSC-NK cells at effectortarget ratios of 1 : 1 and 4:1. Data represented as mean ± SEM.
[0015] FIGs. 9A-J show genetic modification of mature hiPSC-NK cells using lentivirus. hiPSC-NK cells can be transduced with lentiviral -based vectors to express a CD19-CAR construct with high affinity. Conversely, primary NK cells demonstrate a persistent resistance to genetic engineering after lentiviral transduction.
[0016] FIGs. 10 A- J show genetic modification of mature hiPSC-NK cells using retrovirus. Both mature hiPSC-NK and primary NK cells can be transduced with retroviralbased vectors to express a CD19-CAR construct with high affinity.
[0017] FIGs. 11 A-C show comparative functional analysis between unmodified and CAR19 hiPSC-NK against tumor targets. In vitro killing against Raji cells (FIG. 11 A), NALM6 cells (FIG. 1 IB), and CCRF cells (FIG. 11C) as determined using Incucyte’s immune cell-killing software for live cell analysis. Different concentration of target cancer cells was incubated with hiPSC-NK cells or CAR19-hiPSC NK cells at effectortarget ratios of 2: 1. Data represented as mean ± SEM.
[0018] FIGs. 12A-C show genetic modification of iPSCs using lentivirus. FIG. 12A: The selection and colony-picking of a single iPSC-colony from the surface of the cell culture plate. FIG. 12B: After 2 days in differentiation medium, CD19-expression can be observed in the removed colonies under fluorescent imaging. A wide variation in CD19-expressing positive clones derived from selected colonies observed (top and bottom row, yellow arrows and arrowheads). FIG. 12C: After 5 days in differentiation medium, the picked colonies expressing CD 19 are capable of proliferating and maintaining expression in vitro (yellow arrowheads).
[0019] FIGs. 13A-B show NK-differentiation of lenti-CAR19 iPSCs using 3D bioreactor on day 12. FIG. 13A: Flow cytometry quantification for CD34 and CD45 marker expression. The untransduced hiPSC cell line exhibited 15% CD34-positive expression (middle panel) compared to 12% expression in the CAR19 S001 modified hiPSCs (far right panel). Percent positive for CD45 was <1% in both cell lines. FIG. 13B: CD19-CAR expression detected in differentiating spheres in 3D-bioreactor.
[0020] FIG. 14 shows schematics of viral CAR constructs.
[0021] FIGs. 15A-B show that cryopreserved unmodified hiPSC-derived NK cells demonstrate strong killing efficacy in a subcutaneous K562 myelogenous leukemia mouse model. FIG. 15 A: Frozen peripheral blood derived NK (PBNK) or frozen iPSC-derived NK cells were injected intravenously (4 x 106/mouse) post-tumor engraftment. Optical bioluminescence imaging of tumor load between days 3 and 7 post-treatment. FIG. 15B: Quantification of tumor intensity examined across all control and experimental cohorts four days after NK-infusion.
[0022] FIGs. 16A-B show iPSC-derived NK cells can survive and persist in vivo for at least two weeks post-injection. FIG. 16A: Infused NK cells transduced with retroviral constructs expressing luciferase alone or luciferase + soluble IL 15 then were injected intravenously into the tail vein (2 x 106/mouse). FIG. 16B: Persistence of genetically modified iPSC-NK cells were detectable two weeks in vivo. Transduction of IL15 enhanced
persistence of injected iPSC-NK cells, and there were no discernible effects on mice from iPSC-NK infusion.
[0023] FIGs. 17A-D show that NK cells can be generated from a human embryonic stem cell (hESC) source using a 3D-bioreactor platform system. FIGs. 17A-C: High magnification (40x) photomicrographs of cell suspensions collected from the 3D-bioreactor demonstrate a phenotypic maturation of NK cells over developmental time. FIGs. 17D: A purified population of NK cells were collected and maintained over time as indicated by flow cytometry quantification of mature NK cell markers CD45 and CD56.
[0024] FIGs. 18A-F: NK cells generated from a human embryonic stem cell (hESC) source using a 3D-bioreactor platform system show potent endogenous killing activity and can be genetically modified to enhance killing activity using a CAR construct in hepatocellular cancer lines. FIG. 18 A: ESC-CAR NK in vitro killing displayed as percent growth against Huh7 cancer cell line over a 24-hour period. FIG. 18B: Average Huh7 percent cancer growth at 24 hours. FIG. 18C: ESC-CAR NK in vitro killing displayed as percent growth against HepG2 cancer cell line over a 24-hour period. FIG. 18D: Average HepG2 percent cancer growth at 24 hours. FIG.18E: ESC-CAR NK in vitro killing displayed as percent growth against Hep3B cancer cell line over a 24-hour period. FIG. 18F: Average Hep3B percent cancer growth at 24 hours.
DETAILED DESCRIPTION
[0025] A description of example embodiments follows.
Overview
[0026] According to recent statistics provided by the United States Center for Disease Control and Prevention (CDC), deaths from cancer reported within the United States reached approximately 600,000 per year despite advancements in oncological patient care. The development of novel immunotherapies utilizing the potent cytotoxic antitumor properties of Natural Killer (NK) cells are a promising approach in cell-based therapeutics. However, the source of NK cells often poses certain limitations. Since NK cells only compose 5-15% of all lymphocytes circulating in the blood, the isolation of NK cells from the peripheral blood (PBNK) is often inefficient, resulting in a heterogenous lymphocyte population yielding only 10-20% NK cells [1] Additionally, a significant decrease in cell cytotoxicity has been
reported following cryopreservation ([2, 3]). Conversely, NK cells isolated from cord blood (CBNK) exhibit higher cell viability after cryopreservation, however, due to the lower number of NK cells detected per cord blood unit, numerous expansion phases are required to obtain multiple units per dose [4], In comparison to PBNKs, several reports suggest CBNKs display a more immature phenotype that is directly correlated to a reduction in cytotoxicity [4, 5], Although these issues of low NK cell yield and cytotoxicity loss can be partially resolved through cytokine support and feeder cell co-culture, these considerations demonstrate that PBNKs and CBNKs are not ideal for an “off-the-shelf’ allogeneic immunotherapy whereby the NK cells are harvested from unrelated donors, cryopreserved, and thawed.
[0027] Human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs) have the capacity for indefinite self-renewal and the ability to differentiate into all cell types of the body. An infinite starting cell source is a tremendous advantage in scaling-up the production of a homogenous and high-quality NK cell product. This allows for the development and expansion of GMP-grade iPSC-derived NK cells to be used in cancer immunotherapy clinical trials. Several research groups have published recent advances in methodological strategies to successfully differentiate pluripotent stem cells into the NK lineage that display comparable phenotype and function to primary NK cells [6-10], Some of those methods involve differentiation of NK cells by co-culturing with OP9 mouse stromal cells. As human pluripotent stem cells (PSCs) continue to be intensely studied for their potential clinical applications as a cancer treatment strategy, PSC-derived NK cells are a promising alternative approach to cancer immunotherapy.
[0028] As the differentiation of PSCs into the NK specific lineage continues to be extensively investigated, several cell culturing strategies have been utilized to produce high- quality unadulterated NK cells: 1) cytokine induction with feeder-cell co-culture; and 2) the generation of an embryoid body (EB) intermediary with cytokine induction. The application of these different strategies yields varying degrees of success.
[0029] The co-culture of hPSCs with stromal cells, primarily derived from the bone marrow, has been a commonly used method in the induction of the hematopoietic fate by first obtaining a CD34+ precursor population [8, 9, 11], However, the protocols often require a long co-culture period of approximately 21 days in addition to a cell-sorting step for CD34+ positive cell selection before downstream NK differentiation can proceed. The additional cell-sorting component not only results in a less efficient protocol, but it also results in a
decreased NK cell yield by negatively selecting against hematopoietic progenitors necessary for robust NK differentiation.
[0030] The formation of EBs is a commonly used platform in differentiating PSCs to specific cell lineages. However, current methods in EB generation create spheres of unequal size that may lead to irreproducible differentiation results. Additionally, the disintegration of EB-spheres has been observed shortly after the initiation of differentiation. Therefore, the low efficacy and reproducibility in both hematopoietic induction and NK differentiation via the EB step would not be ideal for adaptation into a large scale-production system required in a clinical trial setting.
[0031] Therefore, modifications and optimizations in the biomanufacturing of NK cells for immunotherapies is currently needed[12]. The ability to generate large quantities of GMP-grade NK cells derived from a pluripotent source would prove most useful for cellbased immunotherapies.
[0032] As used herein, pluripotent stem cells (PSC) includes human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs). The methods described here are suitable for use with pluripotent stem cells.
3D Bioreactors
[0033] A three-dimensional (3D) bioreactor promotes growth of cells in three dimensions. A 3D bioreactor is distinguished from two-dimensional (2D) growth of cells on a flat surface, such as a cell culture dish. By promoting cell growth in three dimensions, the cells can grow into spheroids, or 3D cell colonies.
[0034] Some 3D bioreactors have scaffolds to which adherent cells can attach. Other 3D bioreactors employ scaffold-free techniques. One example of a scaffold free 3D bioreactor is a 30 ml disposable magnetic stir bioreactor from ABLE Corporation (Japan) and Biott Corporation (Japan) (ABLE Cat No. BWV-S03 A), which uses an impeller with a magnet on each blade to provide low-shear agitation by laminar flow, which can encourage the formation and growth of spheroid cell clusters. An impeller or other mechanism for mixing the culture media provides multiple benefits, including enhanced mass transfer and increased cell surface area exposure to media, nutrients, and differentiation factors. Relative to a static culture, low-shear stress and fluid flow provide more physiologically relevant environments. Other suitable bioreactor magnetic stir systems are available from ABLE Biott and other manufacturers in a variety of sizes.
[0035] Typically, the 3D bioreactor is an enclosed bioreactor.
Cell culture medium for pluripotent stem cell expansion
[0036] Pluripotent stem cells are cultured in a cell culture medium that maintains them in a pluripotent state as the cells grow from an initial aliquot to a 3D culture. Many commercially-available feeder-free, xenofree culture media are suitable for use as a basal medium for iPSC expansion. One example is Essential 8 medium (available from ThermoFisher Scientific). Others include mTeSR Plus™ (StemCell Technologies);
NutriStem® XF medium (Biological Industries); Cellartis DEF-CS 500 (Takara); and StemFlex™ Medium (Fisher Scientific).
[0037] Another suitable medium is described in Chen, G. et al. Chemically defined conditions for human iPSC derivation and culture. Nat Methods. 2011;8(5):424-429 (Methods, Human ES Cell Culture, E8 media composition). The media described therein includes DMEM/F12, L-ascorbic acid-2-phosphate magnesium (64 mg/1), sodium selenium (14 pg/1), FGF2 (100 pg/1), insulin (19.4 mg/1), NaHCO3 (543 mg/1) and transferrin (10.7 mg/1), TGFpi(2 pg/1) or NODAL (100 pg/1). Osmolarity of all media was adjusted to 340 mOsm at pH 7.4. One of skill in the art will appreciate that the concentrations need not be precisely as described in Chen, G. et al.
[0038] The pluripotent stem cell expansion medium can also include vitronectin, which is a glycoprotein that provides a surface coating to promote cell attachment for use in feeder- free culture of pluripotent stem cells.
[0039] The pluripotent stem cell expansion medium can also include a rho-kinase (ROCK) inhibitor. Typically, a ROCK inhibitor is only used when hiPSCs or hESCs need to be enzymatically passaged to help mitigate cell death. One example of a ROCK inhibitor is Y-27632, which is a compound having the following structure:
[0040] Y-27632 is sometimes available as a salt e.g., ■ 2 HC1).
[0041] After culturing in the pluripotent stem cell expansion medium, the cell density increases and the cells form 3D-PSC spheres after bioreactor adaptation. Expression of OCT- 4, a marker of pluripotency, of the 3D-PSCs can be quantified by flow cytometry. Typically, greater than 90% of the 3D-PSCs resulting from the first culture step express OCT-4.
[0042] In this PSC expansion step, the PSCs are typically cultured to a cell density of at least about 750,000 cells/mL. In some instances, the PSCs are cultured to a cell density from about 750,000 cells/mL to about 1,250,000 cells/mL. For the 30 mL bioreactor used in the Examples, the iPSCs were cultured to approximately 25 million cells to 35 million cells.
[0043] Optionally, the cells can be mechanically dissociated during cell culture (e.g., with a pipette) in order to separate cell clumps or aggregates into smaller clumps, smaller aggregates, or single suspended cells.
Cell culture medium for differentiation of pluripotent stem cells to hematopoietic stem cells (HSCs)
[0044] The cells are then cultured to differentiate the 3D PSCs to hematopoietic stem cells (HSCs).
[0045] Typically, the hematopoietic stem cell culture medium includes stem cell factor (SCF), bone morphogenetic protein 4 (BMP4) (or an agonist of BMP4), and vascular endothelial growth factor (VEGF). The hematopoietic stem cell culture medium typically also includes a basal culture medium that promotes hematopoietic differentiation. One suitable basal culture medium is albumin polyvinylalcohol essential lipids (APEL). Suitable APEL culture media are described in US Patent No. 10,894,944 B2 and in Table 1 of Ng et al., A protocol describing the use of a recombinant protein-based, animal product-free
medium (APEL) for human embryonic stem cell differentiation as spin embryoid bodies, Nature Protocols 3, 768-776 (2008).
[0046] At the conclusion of the hematopoietic stem cell culturing step, about 10-50% of the cells express CD34+, a marker of hematopoietic stem cells (HSCs). Typically, the cells are cultured for about 6 days to about 12 days. After about day 12, the cells begin to transition to a more mature (differentiated) cell type (e.g., natural killer cells). Once the cells are differentiated to HSCs, the cells can be differentiated to natural killer cells.
Cell culture medium for differentiation of HSCs to natural killer cells
[0048] The cells are then cultured to differentiate them from hematopoietic stem cells (HSCs) to a differentiated cell type, such as natural killer cells.
[0049] One example of a cell culture medium includes: i) Dulbecco's Modified Eagle Medium with Glutamax; ii) Dulbecco's Modified Eagle Medium/Nutrient Mixture F-12 with Glutamax containing Human AB serum; iii) ethanolamine; iv) 2-mercaptoethanol (2-BME); v) sodium selenite; and vi) ascorbic acid with interleukin 3 (IL3), interleukin 7 (IL7), interleukin (IL 15), FMS-like tyrosine kinase 3 ligand (Flt3L), stem cell factor (SCF).
[0050] Human AB serum refers to Type AB serum typically derived from male donors. Type AB donors lack antibodies against A and B-blood type antigens, therefore more commonly used to mitigate immunoreactivity.
[0051] Without wishing to be bound by theory, FMS-like tyrosine kinase 3 ligand (Flt3L) can enhance IL15 signaling, which can be important for the NK lineage. IL15 is a cytokine involved in the specific maintenance and proliferation of NK cells, and thus, Flt3L may enhance proliferation and/or NK specification as a result.
[0052] As cells mature and adapt an NK-lineage identity, the NK cells are released into the cell medium, from which they can be collected for use in downstream applications. Cells can be cultured indefinitely in this cell culture medium as NK cells are produced.
Lentiviral transduction
[0053] In some embodiments, the lentivirus is concentrated with Lenti-X Concentrator (Takara Bio, Cat. Nos. 631231 & 631232).
[0054] In some embodiments, lentiviral transduction can be enhanced by culturing with RetroNectin, which is a 63 kD fragment of recombinant human fibronectin fragment (also referred to as rFN-CH-296).
[0055] Lentiviral transduction provides advantages relative to retroviral transduction. Lentiviruses (LVs) can infect non-dividing as well as actively dividing cell types, whereas retroviruses can only infect actively dividing, mitotically active cells. Accordingly, transduction with lentivirus is, in theory, more efficient as it has the potential to incorporate into all NK cells as opposed to only proliferating ones.
[0056] NK cells are resistant to LV transduction, which hampers their development as an immunotherapy. Vesicular Stomatitis Virus type-G (VSV-G) LVs, which are one of the most commonly used viruses for generating chimeric antigen receptor (CAR)-T cells, do not efficiently transduce NK cells.
[0057] While the preferred method of incorporating genetic modifications into iPSC-NKs is via transduction of iPSCs followed by selection of single cell clones to create stable iPSC lines, this is not ideal for high throughput screening purposes as the timeline for stable iPSC generation and subsequent NK differentiation is relatively long.
[0058] Prior studies by others have involved modifying the structure and/or sequence of specific LVs to improve transduction efficiency, but these studies have yielded few promising results. The method described herein of using Lenti-X concentrator in combination with retronectin produces unexpected results in transducing induced natural killer cells (iNKs) with a lentivirus.
[0059] As used herein, the terms “nucleic acid,” “nucleotide,” and “polynucleotide” shall be given their ordinary meanings and shall include deoxyribonucleotides, deoxyribonucleic acids, ribonucleotides, and ribonucleic acids, and polymeric forms thereof, and includes either single- or double-stranded forms. Nucleic acids include naturally occurring nucleic acids, such as deoxyribonucleic acid (“DNA”) and ribonucleic acid (“RNA”) as well as nucleic acid analogs. Nucleic acid analogs include those which include non-naturally occurring bases, nucleotides that engage in linkages with other nucleotides other than the naturally occurring phosphodiester bond or which include bases attached through linkages other than phosphodiester bonds. Thus, nucleic acid analogs include, for example and without
limitation, phosphorothioates, phosphorodithioates, phosphorotriesters, phosphoramidates, boranophosphates, methylphosphonates, chiral-methyl phosphonates, 2-O-methyl ribonucleotides, peptide-nucleic acids (PNAs), locked-nucleic acids (LNAs), and the like. [0060] As used herein, the term “operably linked,” for example in the context of a regulatory nucleic acid sequence being “operably linked” to a heterologous nucleic acid sequence, shall be given its ordinary meaning and shall mean that the regulatory nucleic acid sequence is placed into a functional relationship with the heterologous nucleic acid sequence. In the context of an IRES, “operably linked to” refers to a functional linkage between a nucleic acid sequence containing an internal ribosome entry site and a heterologous coding sequence initiation in the middle of an mRNA sequence resulting in translation of the heterologous coding sequence.
[0061] As used herein, the term “vector” shall be given its ordinary meaning and shall refer to a vehicle by which a DNA or RNA sequence (e.g., a foreign gene) can be introduced into a genetically engineered cell, so as to transform the genetically engineered cell and promote expression (e.g., transcription and/or translation) of the introduced sequence.
Chimeric Antigen Receptors
[0062] Embodiments described herein related to transduction of cells to express a chimeric antigen receptor (CAR). Cells can be transduced by a vector, such as a lentiviral vector, to express a chimeric antigen receptor. For example, a lentiviral vector can include a nucleic acid sequence encoding a chimeric antigen receptor. Chimeric antigen receptors include an extracellular antigen recognition domain, a transmembrane domain, and an intracellular activation domain.
[0063] The extracellular antigen recognition domain can be, for example, a single-chain Fragment variant (scFv) derived from an antibody. In embodiments described herein, the extracellular antigen recognition domain binds CD 19.
[0064] The transmembrane domain can be, for example, a CD3 transmembrane domain, a CD8 transmembrane domain, or a CD28 transmembrane domain.
[0065] The intracellular activation domain can be, for example, a CD3^ domain (sometimes written as CD3-zeta, CD3Z, or CD3z).
[0066] In some embodiments, the chimeric antigen receptor can include a co-stimulatory domain, such as, for example, a 4- IBB domain.
EXEMPLIFICATION
Materials
Cell lines
[0067] hiPSC cell lines were obtained from Al stem that were derived from umbilical cord blood. Human embryonic stem cells (hESCs) were acquired from ESI BIO (Alameda, California, USA). hiPSC culture
[0068] hiPSCs and hESCs were cultured and maintained in Essential 8 medium (ThermoFisher Scientific, Catalog# A2858501). The supplementation of the ROCK Inhibitor Y27632 (Sigma, Cat# SCM075) was added to hiPSC and hESC cultures at lOpM during cell passaging only and removed from the medium within 24 hours of plating.
Hematopoietic stem cell induction medium
[0069] Medium composed of STEMdiff APEL 2 (albumin, polyvinyl alcohol, essential lipids; Stemcell Technologies, Cat# 05275) basal cell culture medium supplemented with Stem Cell Factor (R&D Systems, SCF, 40ng/ml, Cat# 255-SC/CF-050), Bone Morphogenetic Protein 4 (R&D Systems, BMP4, 20ng/ml, Cat# 314-BP-050) and Vascular Endothelial Growth Factor (R&D Systems, VEGF, 20ng/ml, Cat# 293-VE-050).
NK cell differentiation medium
[0070] Basal medium is composed of a mixture of 56.6% DMEM + Glutamax (Life Technologies, Cat# 10566-016) and 28.3% F-12 + Glutamax (Life Technologies, Cat# 31765035) supplemented with 15% human heat-inactivated type-AB serum (Valley Biomedical, Cat# HP1022 HI), 50pM Ethanolamine (MP Biomedicals, Cat# 194658), 25pM 2-Mercaptoethanol (Sigma, Cat# M6250), 5ng/ml sodium selenite (Sigma, Cat# S5261), 20mg/ml ascorbic acid (Sigma, Cat# A-5960), and 1% penicillin-streptomycin (Gibco, Cat# 15140-148). Cytokine addition into the medium consisted of 5ng/ml IL-3 (Peprotech, Cat# 200-03), 20ng/ml IL-7 (Peprotech, Cat# 200-07), lOng/ml IL-15 (Peprotech, Cat# 200-15), lOng/ml Flt3 ligand (Peprotech, Flt3L, Cat# 300-19), and 20ng/ml SCF.
Nalm-6, Raji, and Jurkat cancer cells
[0071] Maintained in Stem Cell Growth Medium (CellGenix, SGCM, Cat# 20802-0500) with 10% fetal bovine serum (Gibco, FBS, Cat# 10082-147), and 1% penicillin/ streptomycin.
Other materials
[0072] TrypLE Select (Life Technologies, Cat# 12563011), Accutase (Innovative Cell Technologies, Inc., Cat# AT105-500, Vitronectin (ThermoFisher Scientific, Cat# A14700), Retronectin (Takara, Cat# T100A), Dnase 1 (Worthington Biochemical, Cat# LK003172), 70pm (Fisherbrand, Cat#22363548), Incucyte Nuclight Red Lentivirus Reagent (EFN-alpha; Puro; Cat#4625), 96 well assay plate (Corning, Cat#3610), 30 ml disposable bioreactor (Able Biott, Cat# ABBWVS03A-6).
Methods
Feeder-free pluripotent stem cell culture and maintenance
[0073] Undifferentiated feeder-free hiPSCs and hESCs are cultured on vitronectin coated substrates in Essential 8 medium. When hiPSCs and hESCs cultures are >70% confluent, cells can be enzymatically passaged using either TrypLE or Accutase and re-seeded in a split ratio between 1 : 10 and 1 : 15 with Y27632 supplementation at a final concentration of lOpM. hiPSCs and hESCs can be continually maintained, expanded, or cryopreserved under these outlined conditions. Prior to initiating 2D-monolayer to 3D-bioreactor hiPSC adaptation, hiPSCs and hESCs need to be expanded until sufficient cells needed have been obtained.
3D-bioreactor adaptation
[0074] hiPSCs and hESCs are transferred as a starting 2D-monolayer into small-scale bioreactors to initiate 3D-suspension culture. When 2D-hiPSCs and hESCs expansion has yielded a cell concentration of more than about 30 x 106 live cells, the undifferentiated hiPSCs and hESCs can be dissociated with TrypLE or Accutase. Cell contents should be filtered through a 70pM cell strainer to remove doublets.
[0075] Optimal seeding density into bioreactors should be approximately 1 x 106 cells/ml in Essential 8 medium with Y27632 supplementation at a final concentration of 10 pM. The rotational speed of the bioreactor at all phases of the 3D-differentiation process ranges from 60-80 RPM. Eighteen to twenty-four hours after adaptation, spheres should be examined carefully under the microscope and assessed for viability and morphology.
Hematopoietic lineage induction
[0076] As illustrated in FIG. 1, the induction of pluripotent stem cells (hiPSCs and hESCs) towards a hematopoietic fate can be initiated after successful 3D- adaptation performed from the previous day. The complete removal of Essential 8 medium from the suspension culture is critical and can be executed by transferring the entire volume from the bioreactor to a 50ml conical tube. Leave cells undisturbed for 10-12 minutes to let the spheres pellet to the bottom. Remove the cell supernatant leaving the pellet undisturbed and replace with Hematopoietic induction medium described previously. Prepare the induction medium by formulating Stemdiff APEL 2 medium supplemented with SCF at a concentration of 40ng/ml in conjunction with BMP4 and VEGF at a concentration of 20ng/ml each. Medium change and preparation can be conducted as described during hematopoietic induction stage between differentiation days 1 to 12.
[0077] The expression of the hematopoietic stem cell progenitor marker CD34 and mature NK marker CD45 can be quantified at Day 6 and Day 12 to analyze the progression of NK cell development over time.
NK cell differentiation
[0078] After twelve days in HSC induction medium, the 3D-sphere aggregates can be further differentiated into the NK cell lineage by replacing the cultures with the above- mentioned NK cell differentiation medium. The NK cell differentiation base medium is composed of 56.6% DMEM + Glutamax and 28.3% F-12 + Glutamax supplemented with 15% human heat-inactivated type- AB serum, 50pM Ethanolamine, 25 pM 2- Mercaptoethanol, 5ng/ml sodium selenite, 20mg/ml ascorbic acid, and 1% penicillinstreptomycin. Cytokine addition into the medium consisted of 5ng/ml IL-3 (Peprotech, Cat# 200-03), 20ng/ml IL-7, lOng/ml IL- 15, lOng/ml Flt3 ligand, and 20ng/ml SCF. From differentiation day 12 and onwards, only NK differentiation medium is required. Medium in the 3D-bioreactor cultures should be replaced daily or when needed.
[0079] Typically, pluripotent stem cell-derived NK cells delaminate from the differentiating 3D-sphere aggregates and are released into the cell culture medium suspension after detachment. Mature appearing NK cells can be observed floating in the cell medium after 1 week of NK cell differentiation. hiPSC derived NK cells can be collected directly from the medium and phenotyped for surface antigens that include CD45, CD56, CD 16, KIR,
CD94, NKp44, NKG2D, and Lampl using flow cytometry. Functional analyses of hiPSC- derived NK cells can be assessed by in vitro killing assays against numerous cell cancer lines at various effector to target (E:T) ratios. NK cells developed using this 3D-bioreactor platform demonstrate a mature NK cell lineage profile and potent cell cytotoxicity.
[0080] The methods outlined indicate an efficient and streamlined approach in deriving mature and functional NK cells from a pluripotent stem cells source that can be used as an infinite cell source for potential cancer cell therapies.
In vitro killing assay
[0081] The cancer target cells are labeled with the Incucyte Nuclight Red lentivirus, which enables the counting of viable cancer cells over time. Puromycin is added to the cancer cell cultures on day 6 at a concentration of 0.5ug/ml to initiate the selection process. Monitor transduction efficiency daily and supplement with puromycin into the cell culture medium with every media change. By day 10-14, a high percentage of the cancer target cells should be Nuclight red.
[0082] Both effector and target cells are cultured in RPMI medium supplemented with 10% FBS for cytotoxicity analysis. Real-time NK cell cytotoxicity was monitored and quantified using Incucyte Base Analysis Software.
Generation and purification of retrovirus for transduction of iPSC-NK
[0083] Plate 3 x 106 293T cells in a 10 cm dish in 10 ml DMEM medium. Prepare two sterile Eppendorf tubes for each dish. In tube 1, add 420ul DMEM to a sterile Eppendorf tube add 30 pl of FugeneHD directly to the DMEM in the Eppendorf tube. Gently tap the tube to mix and incubate for 5 minutes at room temperature (RT). In tube 2, add plasmid(s) to a combined total lOug DNA. Dropwise add the FugeneHD/DMEM mixture to the tube containing the DNA. Gently tap the tube to mix the FugeneHD/DMEM/DNA mixture, and incubate for 15 minutes at RT. Dropwise, add the FugeneHD/DMEM/DNA solution to the cell with the gentle agitation. Incubate cells at 37oC for 48 hours. Harvest the supernatant and centrifuge at 400x g for 5 minutes and store in 4oC. Filter the supernatant with 0.45 um low binding filter. For long term storage, store as 3 or 6 ml aliquots and at -80°C
Virus transduction of iPSC-NK expanded under feeder-free conditions
[0084] Harvest the virus-containing supernatants and filter through a 0.45 pm low binding filter. Transfer supernatant to a sterile container and combine 1 volume of Lenti-X Concentrator with 3 volumes of clarified supernatant and mix by gentle inversion. Incubate mixture at 4°C overnight. Centrifuge sample at 1,500 x g for 45 minutes at 4°C. Carefully remove supernatant and gently resuspend the pellet in 1/10 to 1/100th of the original volume using complete media. Immediately titrate sample or store at -70°C in single-use aliquots. Coat 24 well non-tissue culture treated plate with Retronectin (7ug/mL in PBS). The following day, aspirate Retronectin and wash wells with complete RPMI media. Add 2ml of concentrated virus to Retronectin-coated wells. Spin plate at 2000 g for at least 1 hr at room temperature. Prepare NK MACs media with 500IU/ml IL-2 and 140IU/ml IL-15. Add 2.5E5 cells/ml (1ml per 24 well). Culture undisturbed for a minimum of 2 days before subsequent assays. Monitor CAR expression using flow cytometry and use transduced iNKs for subsequent killing assays.
In vivo killing assay
[0085] Subcutaneous injections of K562 cancer cells were prepared at 2 x 105 cells/mouse. K562 cancer cells were allowed to engraft for 3 days before systematic infusion of hiPSC-NK cells. The cryopreserved hiPSC-NK cells were thawed and prepared for infusion on the same day. Sample preparation for both hiPSC-NK and PBNK were identical as 4 x 106 NK cells/mouse were intravenously injected. Intraperitoneal (IP) injections of recombinant human IL-15 at 30 ng/mouse was administered daily for days 1-7 after NK cell infusion.
Results and Discussion
3D-bioreactor adaptation and iPSC-NK differentiation protocol
[0086] As illustrated in FIG. 1, which is a summary of a 3D-directed in vitro differentiation protocol for NK cells derived from human induced pluripotent stem cells (hiPSC), the induction of hiPSCs towards a hematopoietic fate can be initiated after successful 3D-hiPSC adaptation, which requires approximately 12 days, after which time 3D- cultures are transferred to NK cell differentiation conditions. Two weeks thereafter, mature and functional NK cells can be continuously collected from the medium for a period of about 35 days. Briefly, hiPSCs were scaled-up from a 2D-monolayer to a 3D-small scale bioreactor
cell culture system with Y27632 supplementation to enhance iPSC viability and improve sphere formation.
[0087] hiPSCs were grown as 2D-monolayer cultures to a starting concentration of 30 x 106 live cells. These undifferentiated hiPSCs were dissociated with TrypLE then filtered through a 70pM cell strainer to achieve single cell suspension. The 3D-adaptation of hiPSCs from 2D-monolayer culture was completed utilizing a small-scale bioreactor. FIG. 2A is a low magnification (lOx) photomicrograph of hiPSCs in 2D-monolayer culture. FIG. 2B is a photograph of a small-scale 30 ml bioreactor flask. FIG. 2C is a photomicrograph of hiPSC- spheres 20-hours after 2D-to-3D adaptation.
Phenotypic characterization of 3D-bioreactor-derived hiPSC-NK cells
[0088] Importantly, methods of hiPSC-NK differentiation have been described using adherent 2D-monolayer cultures. The 3D differentiation protocol described herein was compared to a traditional method of adherent 2D hiPSC-NK differentiation.
[0089] In comparing the differentiation potential between 2D-monolayer versus 3D- bioreactor generated hiPSC-NK cells, NK cells were observed and specified in medium suspension almost 10 days faster when compared to 2D-controls (FIGs. 3A-H). FIGs. 3A-D are images of adherent-2D derived cell-spheres and derivatives at indicated time points after the initiation of NK differentiation (lOx). Spheres slowly flatten over time with premature NK-like cells observed in cell medium at differentiation day 23. FIGs. 3E-H are images of 3D bioreactor generated cell-spheres and derivatives. Immature NK-like cells are observed two days after NK differentiation initiation on day 14.
[0090] In addition, 3D-bioreactor-derived NK cells demonstrate advanced maturation and development. Flow cytometry quantification indicates a correlated temporal pattern of expression for the hematopoietic stem cell progenitor marker CD34 (FIG. 4A) and NK cell markers CD45 and CD56 (FIGs. 4B-F). Expression levels of CD56, a marker for mature NK cells, demonstrated that the 3D bioreactor-derived cells were over 93% positive at day 27 of differentiation compared to 58% at day 27 of differentiation for 2D-generated cultures (FIGs. 5A-F and 5). Comparison of expression levels of CD56 over time reveals that bioreactor- derived cells express consistently higher levels of CD56 across all timepoints tested as compared to similarly staged 2D samples (FIG. 5).
[0091] To further assess the robustness and timing of cellular differentiation between the 3D-bioreactor and 2D-monolayer platforms, expression levels were measured for NK
lineage-specific markers CD94, NKp44, NKG2D, Lampl, and CD 16. Expression levels of these NK lineage markers for differentiation day 27 bioreactor-generated cells were comparable or significantly exceed expression levels from two independent 2D- differentiation rounds that were cultured almost twice as long (53 days and 55 days, respectively) (FIG 6, and Table 1).
Table 1. Differential expression of lineage-specific surface markers comparing 2D versus 3D differentiation methods.
[0092] As an additional comparison, 3D-bioreactor-derived hiPSC-NK cells were compared to primary cord blood-derived NK (CBNK) cells (FIGs. 7A-J). hiPSC-NK cells expressed similar levels of mature NK markers CD56, CD 16, CD94, NKG2D, NKp44 and LAMP1 as compared to primary CBNK cells. Culturing hiPSCs in a 3D bioreactor using the methods and culture media described herein can generate highly pure and phenotypically mature cells that are similar to primary NK cells.
Functional in vitro killing capacity of 3D-bioreactor-derived hiPSC-NK cells
[0093] While it is important that NK cells express appropriate maturation markers, the gold standard for validating the phenotype of these cells is by assessing the functional killing capacity of the final NK product. Functional analyses of hiP SC -derived NK cells were
assessed by in vitro killing assays against Jurkat (FIG. 8A), HeLa (FIG. 8B), and K562 (FIG. 8C) cancer cells as determined using using Incucyte Base Analysis immune killing software for live cell analysis. Different concentrations of target cancer cells were incubated with hiPSC-NK cells at effectortarget (E:T) ratios of 1 : 1 and 4: 1. For additional comparison, similar killing assays were performed to compare the functionality of hiPSC-NK to primary CBNK at an E:T ratio of 1 :2. Importantly, hiPSC-NK performed as well as (if not better than) CBNK in K562 killing (FIG. 8D). Thus, NK cells developed using this 3D-bioreactor platform demonstrate a mature NK cell lineage profile and potent cell cytotoxicity.
Virus transduction of pre-differentiated hiPSC-NK expanded under feeder-free conditions [0094] One method for incorporating genetic modifications into iPSC-NKs is via transduction of iPSCs followed by selection of single cell clones to create stable iPSC lines. However, this method is not ideal for high throughput screening purposes as the timeline for stable iPSC generation and subsequent NK differentiation is relatively long. The transduction protocol described herein was used to incorporate both lentiviral and retroviral constructs into pre-differentiated hiPSC-NK cells.
[0095] Genetic modification of NK cells can be performed to improve their function, target specific antigens, and/or enhance persistence in vivo. One application in the development of NK therapeutics is the incorporation of different chimeric antigen receptor (CAR) constructs. Introducing CAR constructs into NK cells has been hindered by the relative difficulty of viral modification of NK cells in comparison with T cells. Use of retroviral vectors for NK transduction has become more common. As retroviruses specifically infect proliferating cells, substantial efforts have been made to enhance NK proliferation as a means of enhancing transduction efficiency. In comparison with retroviruses, lentiviral vectors can be advantageous because they reduce the potential for genomic insertion and potential off-target effects. However, transduction of NK cells with lentiviral vectors has been significantly more challenging than transduction with retroviral vectors.
[0096] Differentiated, mature hiPSC-NK cells as well as primary adult peripheral blood- derived NK (PBNK) cells were transduced with either lentivirus (FIGs. 9A-J) or retrovirus (FIGs. 10A-J) using the protocol described herein. hiPSC-NK cells can be transduced with both retroviral and lentiviral -based vectors to express a CD19-CAR construct with high affinity. Conversely, primary PBNK cells demonstrate a persistent resistance to lentiviral transduction. Schematics of the constructs are depicted in FIG. 14.
Functional in vitro killing capacity of pre-differentiated hiPSC-NK cells transduced with CAR19
[0097] 3D-bioreactor derived hiPSC-NKs were used to transduce a retroviral construct expressing both CAR19 as well as an IL- 15 overexpression cassette using the transduction method described herein. Resulting CAR19 expression was approximately 47%. Nontransduced (NT) or CAR19 hiPSC-NKs were subjected to an in vitro killing assay against CD19+ Raji cells (FIG. 11 A), CD19+ NALM6 cells (FIG. 1 IB), and CD19-CCRF cells (FIG. 11C) as determined using Incucyte’s immune cell-killing software for live cell analysis. Different concentrations of target cancer cells were incubated with hiPSC-NK cells or CAR19-hiPSC NK cells at effectortarget ratios of 2: 1. Importantly, CAR19-transduced hiPSC-NK cells showed enhanced killing efficiency against cancer cell lines expressing the CD 19 target.
Lentivirus transduction of undifferentiated hiPSCs
[0098] A preferred method of incorporating genetic modifications into iPSC-NKs is via transduction of iPSCs followed by selection of single cell clones to create stable iPSC lines. Stably introducing CAR constructs at the iPSC-stage, allows for the persistence of this genetic modification in all differentiated progenitors, resulting in an ultimate CAR transduction efficiency of 100%. Importantly, some genetic modifications induced during the iPSC-stage may result in impaired differentiation into the desired cell type. As proof of principle that genetic modifications performed at the iPSC-stage do not impair 3D bioreactor- mediated NK differentiation, lentiviral transduction was performed on iPSC colonies, which were subsequently expanded as undifferentiated iPSCs for subsequent differentiation within the enclosed bioreactor system (FIGs. 12A-C). Single colonies from iPSC cultures were selected and replated onto new vitronectin substrates in the presence of concentrated lentivirus expressing a CAR19 construct (FIG. 12 A). After 2 days in differentiation medium, CAR19-expression can be observed in the removed colonies under fluorescent imaging. As the selected colonies propagate over time, an expansion of CAR19-expressing cells is observed, as indicated by the binding of CD19-GFP proteins to iPSCs successfully infected with CAR19. This CAR construct is retained after numerous rounds of symmetric cell division (FIGs. 12B-C). Without wishing to be bound by theory, due to the epigenetic silencing of introduced constructs that naturally occurs during pluripotency, the expression of
this CAR construct is variable due to methylation of non-pluripotent genes. After 5 days in differentiation medium, the picked colonies expressing CAR19 are capable of proliferating and maintaining expression in vitro. Using this method demonstrates that most progeny of CAR19-transduced iPSCs express the induced CAR19 construct.
3D bioreactor-based differentiation of lenti-CAR19 hiPSCs
[0099] After generating CAR19-hiPSCs via single colony transduction, both nontransduced (NT) and CAR19 hiPSC lines were induced to differentiate into NK cells using the 3D bioreactor protocol. This approach allowed for the continued monitoring of the response of genetically modified hiPSC lines to the previously established 3D bioreactor differentiation protocol. As such, separate bioreactors were used to differentiate both NT and CAR19 hiPSC lines. On day 12, hematopoietic differentiation as determined by CD34 and CD45 expression were unchanged between NT and CAR19 hiPSC lines (FIG. 13 A). NT hiPSC cell line exhibited 15% CD34-positive expression (FIG. 13 A, middle panel) compared to 12% expression in the CAR19 S001 modified hiPSCs (FIG. 13A, right panel). Percent positive for CD45 was <1% in both cell lines. CAR19-transduced hiPSC lines were assayed for CAR19 expression (FIG. 13B), which demonstrated CD19-CAR expression in differentiating spheres in 3D-bioreactor.
Post-differentiation expansion of iPSC-NK
[00100] In comparison to other known methods, the methods described herein are performed under xeno-free and feeder-free conditions. Consequently, the final derived cells are GMP-compliant and can be used in, e.g., clinical trials. One small-scale (30 ml) bioreactor can potentially yield >10 x 107 to 50 x 107 total NK cells. This refinement of this hiPSC to NK cell differentiation process allows the opportunity to scale-up enough highgrade cells to meet the stringent requirements needed to proceed with human clinical trials. Additionally, the methods described herein are efficient and cost-effective for generating phenotypically mature, functional, clinical-grade NK cells for use in developing cancer immunotherapies.
Phenotypic characterization and killing efficacy of 3D-bioreactor derived embryonic stem cell (ESC)-NK cells
[00101] Embryonic stem cells (ESCs) also generated NK cells at a similar relative efficiency to iPSCs following the differentiation protocol in the 3D-bioreactor platform system as previously described. A purified population of NK cells that express mature NK lineage markers were collected in suspension from the bioreactor medium (FIGs. 17A-D). These ESC-NK cells demonstrate robust in vitro killing against HUH7, HEPG2, and HEP3B cancer cell lines compared to the PBNK-control (FIGs. 18A-F). ESC-NK cells genetically modified to express a CAR (ESC-NK CAR) demonstrated a greater reduction in HUH7, HEPG2, and HEP3B cancer cells compared to non-transformed ESC-NK cells (ESC-NK NT).
Functional in vivo killing efficacy of 3D-bioreactor-derived hiPSC-NK cells in a myelogenous K562 cancer mouse model
[00102] To assess the killing efficacy of the iPSC-derived NK cells in vivo, cryopreserved hiPSC-NK cells were thawed and prepared the same day for infusion into K562 xenografted mice. The K562 cancer line was injected subcutaneously and allowed to engraft for three days before a single dose of hiPSC-NK cells was injected intravenously. Bioluminescent imaging four days post-NK injection showed significant tumor regression in the hiPSC-NK mice in comparison to the PBNK and PBS control groups (FIG. 15A-B). Additionally, the hiPSC-NK injected mice demonstrated slower tumor growth in comparison to the PBNK and PBS control groups as illustrated by luciferase expression (FIG. 15B). Importantly, this illustrates that the hiPSC-NK cells derived using the 3D-bioreactor system are functionally potent and capable of short-term anti-tumor efficacy in vivo.
In vivo persistence of hiPSC-NK genetically modified to express IL-15
[00103] To assess the persistence of hiPSC-NK in vivo, post-differentiated hiPSC-NK cells were transduced with retroviral constructs expressing luciferase alone or luciferase + secreted IL15, using methods described herein. Transduced hiPSC-NK cells were intravenously infused via tail vein injection (2 x 106/mouse) then tracked over time using bioluminescent imaging. These genetically modified hiPSC-NK cells persisted in vivo for up to two weeks (FIGs. 16A-B). Those hiPSC-NK cells transduced with IL- 15 demonstrated
enhanced persistence in vivo. Importantly, there were no discernible effects on mice resulting from the hiPSC-NK infusion.
REFERENCES
[00104] 1. Koepsell, S .A., et al., Successful ,Auin-flight,Au activation of natural killer cells during long-distance shipping. Transfusion, 2013. 53(2): p. 398-403.
[00105] 2. C, M., et al., - Cryopreservation impairs 3-D migration and cytotoxicity of natural killer cells. - Nat Commun. 2020 Oct 16; 11(1):5224. doi: 10.1038/s41467-020- 19094-0., (- 2041-1723 (Electronic)): p. T - epublish.
[00106] 3. Szmania, S., et al., Ex vivo-expanded natural killer cells demonstrate robust proliferation in vivo in high-risk relapsed multiple myeloma patients.
[00107] 4. Wang, H., et al., The unexpected effect of cyclosporin A on CD56+CD16- and
CD56+CD16+ natural killer cell subpopulations. (0006-4971 (Print)).
[00108] 5. Luevano, M., A. Madrigal, and A. Saudemont, Generation of natural killer cells from hematopoietic stem cells in vitro for immunotherapy. Cell Mol Immunol, 2012. 9(4): p. 310-20.
[00109] 6. Hermanson DI and Kaufman Ds, Utilizing chimeric antigen receptors to direct natural killer cell activity. (1664-3224 (Print)).
[00110] 7 Hermanson, D.L., et al., Induced Pluripotent Stem Cell-Derived Natural Killer
Cells for Treatment of Ovarian Cancer.
[00111] 8. Knorr, D.A., et al., Clinical-scale derivation of natural killer cells from human pluripotent stem cells for cancer therapy.
[00112] 9. Woll, P.S., et al., Human embryonic stem cells differentiate into a homogeneous population of natural killer cells with potent in vivo antitumor activity.
[00113] 10. Ni, Z., et al., Expression of Chimeric Receptor CI)4(E by Natural Killer
Cells Derived from Human Pluripotent Stem Cells Improves In Vitro Activity but Does Not Enhance Suppression of HIV Infection In Vivo. Stem Cells, 2014. 32(4): p. 1021-1031.
[00114] 11. Woll, P.S., et al., Human embryonic stem cell-derived NK cells acquire functional receptors and cytolytic activity.
[00115] 12. Shankar, K., C.M. Capitini, and K. Saha, Genome engineering of induced pluripotent stem cells to manufacture natural killer cell therapies. Stem Cell Research & Therapy, 2020. 11(1): p. 234.
[00116] 13. Chen, G. et al. Chemically defined conditions for human iPSC derivation and culture. Nat Methods. 2011;8(5):424-429 (Methods, Human ES Cell Culture, E8 media composition).
INCORPORATION BY REFERENCE; EQUIVALENTS
[00117] The teachings of all patents, published applications and references cited herein are incorporated by reference in their entirety.
[00118] While example embodiments have been particularly shown and described, it will be understood by those skilled in the art that various changes in form and details may be made therein without departing from the scope of the embodiments encompassed by the appended claims.
Claims
1. A method of making hematopoietic stem cells, the method comprising: a) culturing pluripotent stem cells in a three-dimensional (3D) bioreactor in a first cell culture medium to produce a 3D culture of pluripotent stem cells having a cell density of at least about 750,000 cells/mL; and b) culturing the 3D culture of pluripotent stem cells in the three-dimensional (3D) bioreactor in a second cell culture medium to produce a 3D culture of hematopoietic stem cells, wherein the second cell culture medium comprises: i) stem cell factor (SCF); ii) bone morphogenetic protein 4 (BMP4) or an agonist of BMP4; and iii) vascular endothelial growth factor (VEGF).
2. The method of claim 1, wherein a) comprises producing a three-dimensional culture of pluripotent stem cells having a cell density from about 750,000 cells/mL to about 1,250,000 cells/mL.
3. The method of claim 1, wherein b) comprises culturing for at least six days.
4. The method of claim 1, wherein b) comprises culturing for six days to fourteen days.
5. The method of claim 1, wherein the pluripotent stem cells are induced pluripotent stem cells.
6. The method of claim 5, wherein the induced pluripotent stem cells are human induced pluripotent stem cells.
7. The method of claim 1, wherein the pluripotent stem cells are embryonic stem cells.
8. The method of claim 7, wherein the embryonic stem cells are human embryonic stem cells.
9. The method of claim 1, wherein the first cell culture medium comprises a rho-kinase (ROCK) inhibitor.
- 26 -
The method of claim 9, wherein the ROCK inhibitor is Y27632. The method of claim 1, wherein the 3D bioreactor is a scaffold-free bioreactor. The method of claim 11, wherein the scaffold-free bioreactor is a rotational bioreactor. The method of claim 12, wherein the rotational bioreactor rotates at a speed from 60 revolutions per minute to 80 revolutions per minute. The method of claim 1, wherein the hematopoietic stem cells are CD34+. The method of claim 1, wherein the concentration of stem cell factor is from about 5 ng/ml to about 100 ng/ml. The method of claim 1, wherein the concentration of BMP4 is from about 5 ng/ml to about 100 ng/ml. The method of claim 1, wherein the concentration of VEGF is from about 5 ng/ml to about 100 ng/ml. The method of claim 1, wherein the second culture medium further comprises: iv) albumin polyvinylalcohol essential lipids (APEL). The method of any one of claims 1 through 18, further comprising: c) culturing the hematopoietic stem cells in the 3D bioreactor in a third cell culture medium to produce natural killer (NK) cells. The method of claim 19, further comprising transducing the NK cells with a lentiviral vector. The method of claim 20, wherein the lentiviral vector comprises a nucleic acid sequence encoding a chimeric antigen receptor. The method of claim 21, wherein the chimeric antigen receptor comprises an extracellular antigen recognition domain, a transmembrane domain, and an intracellular activation domain.
The method of claim 22, wherein the extracellular antigen recognition domain is a single-chain Fragment variant (scFv) derived from an antibody. The method of claim 22, wherein the intracellular activation domain is a CD3(^ domain. The method of claim 22, wherein the chimeric antigen receptor further comprises a co-stimulatory domain. The method of claim 25, wherein the co-stimulatory domain is a 4-1BB domain. The method of claim 20, wherein transducing the NK cells with a lentiviral vector comprises culturing with retronectin. The method of claim 19, further comprising quantifying cells expressing CD56, and when at least 75% of the cells express CD56, cryopreserving the NK cells. The method of any one of claims 19 through 28, wherein the third cell culture medium comprises: i) 2-mercaptoethanol (2-BME); ii) sodium selenite; iii) ascorbic acid; iv) interleukin 3 (IL-3); v) interleukin 7 (IL-7); vi) interleukin (IL- 15); vii) FMS-like tyrosine kinase 3 ligand (FLT3L); and viii) stem cell factor (SCF). The method of claim 29, wherein the concentration of 2-BME is from about 5 pM to about 100 pM. The method of claim 29, wherein the concentration of sodium selenite is from about 0.5 ng/ml to about 50 ng/ml. The method of claim 29, wherein the concentration of ascorbic acid is from about 2 pg/ml to about 200 pg/ml.
The method of claim 29, wherein the concentration of IL-3 is from about 0.5 ng/ml to about 50 ng/ml. The method of claim 29, wherein the concentration of IL-7 is from about 2 ng/ml to about 200 ng/ml. The method of claim 29, wherein the concentration of IL-15 is from about 2 ng/ml to about 200 ng/ml. The method of claim 29, wherein the concentration of FLT3L is from about 2 ng/ml to about 200 ng/ml. The method of claim 29, wherein the concentration of stem cell factor is from about 2 ng/ml to about 200 ng/ml. The method of claim 29, the third cell culture medium further comprises ethanolamine. The method of claim 38, wherein the concentration of ethanolamine is from about 5 pM to about 500 pM. A cell culture medium comprising: a) stem cell factor (SCF); b) bone morphogenetic protein 4 (BMP4) or an agonist of BMP4; and c) vascular endothelial growth factor (VEGF). The cell culture medium of claim 40, wherein the cell culture medium further comprises: d) albumin polyvinylalcohol essential lipids (APEL).
- 29 -
Priority Applications (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN202280055442.9A CN117836405A (en) | 2021-08-13 | 2022-08-11 | Three-dimensional culture of pluripotent stem cells to produce hematopoietic stem cells |
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US202163232911P | 2021-08-13 | 2021-08-13 | |
US63/232,911 | 2021-08-13 |
Publications (2)
Publication Number | Publication Date |
---|---|
WO2023019213A1 true WO2023019213A1 (en) | 2023-02-16 |
WO2023019213A8 WO2023019213A8 (en) | 2023-03-30 |
Family
ID=85200979
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2022/074853 WO2023019213A1 (en) | 2021-08-13 | 2022-08-11 | Three-dimensional culturing of pluripotent stem cells to produce hematopoietic stem cells |
Country Status (2)
Country | Link |
---|---|
CN (1) | CN117836405A (en) |
WO (1) | WO2023019213A1 (en) |
Citations (7)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2014165131A1 (en) * | 2013-03-13 | 2014-10-09 | Wisconsin Alumni Research Foundation | Methods and materials for hematoendothelial differentiation of human pluripotent stem cells under defined conditions |
WO2017075389A1 (en) * | 2015-10-30 | 2017-05-04 | The Regents Of The Universtiy Of California | Methods of generating t-cells from stem cells and immunotherapeutic methods using the t-cells |
WO2017078807A1 (en) * | 2015-11-04 | 2017-05-11 | Fate Therapeutics, Inc. | Methods and compositions for inducing hematopoietic cell differentiation |
WO2020086889A1 (en) * | 2018-10-24 | 2020-04-30 | Hebecell Corporation | Methods and systems for manufacturing hematopoietic lineage cells |
US10654928B2 (en) * | 2012-10-02 | 2020-05-19 | Memorial Sloan-Kettering Cancer Center | Compositions and methods for immunotherapy |
US20200297763A1 (en) * | 2018-10-18 | 2020-09-24 | Zhejiang University | Pluripotent stem cell-derived macrophage capable of targeting tumor cells and preparation method thereof |
WO2021107779A1 (en) * | 2019-11-28 | 2021-06-03 | Glycostem Therapeutics B.V. | Method for obtaining car-nk cells |
-
2022
- 2022-08-11 CN CN202280055442.9A patent/CN117836405A/en active Pending
- 2022-08-11 WO PCT/US2022/074853 patent/WO2023019213A1/en active Application Filing
Patent Citations (7)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US10654928B2 (en) * | 2012-10-02 | 2020-05-19 | Memorial Sloan-Kettering Cancer Center | Compositions and methods for immunotherapy |
WO2014165131A1 (en) * | 2013-03-13 | 2014-10-09 | Wisconsin Alumni Research Foundation | Methods and materials for hematoendothelial differentiation of human pluripotent stem cells under defined conditions |
WO2017075389A1 (en) * | 2015-10-30 | 2017-05-04 | The Regents Of The Universtiy Of California | Methods of generating t-cells from stem cells and immunotherapeutic methods using the t-cells |
WO2017078807A1 (en) * | 2015-11-04 | 2017-05-11 | Fate Therapeutics, Inc. | Methods and compositions for inducing hematopoietic cell differentiation |
US20200297763A1 (en) * | 2018-10-18 | 2020-09-24 | Zhejiang University | Pluripotent stem cell-derived macrophage capable of targeting tumor cells and preparation method thereof |
WO2020086889A1 (en) * | 2018-10-24 | 2020-04-30 | Hebecell Corporation | Methods and systems for manufacturing hematopoietic lineage cells |
WO2021107779A1 (en) * | 2019-11-28 | 2021-06-03 | Glycostem Therapeutics B.V. | Method for obtaining car-nk cells |
Also Published As
Publication number | Publication date |
---|---|
CN117836405A (en) | 2024-04-05 |
WO2023019213A8 (en) | 2023-03-30 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
AU2020264375B2 (en) | Method for developing natural killer cells from stem cells | |
KR102575976B1 (en) | Proliferation method of natural killer cells | |
AU2007238660C1 (en) | Hemangio-colony forming cells | |
JP2024026891A (en) | Methods and systems for manufacturing hematopoietic lineage cells | |
KR20110006705A (en) | Methods for producing enucleated erythroid cells derived from pluripotent stem cells | |
JP2021072779A (en) | Method for creating t cell precursor cell | |
JPWO2018143243A1 (en) | Method for producing induced pluripotent stem cells | |
US12037607B2 (en) | Methods of preparing hematopoietic progenitor cells in vitro | |
JP7098187B2 (en) | Pluripotent stem cells and their differentiated T cells and their use | |
WO2023019213A1 (en) | Three-dimensional culturing of pluripotent stem cells to produce hematopoietic stem cells | |
JP2023504075A (en) | Method for obtaining CAR-NK cells | |
WO2023243627A1 (en) | Thymic epithelial cell production method | |
US20230149464A1 (en) | Feeder free cell culture methods for expanding natural killer cell preparations | |
EP3564363A1 (en) | Production of megakaryocytes in bioreactors | |
WO2024073665A1 (en) | Scalable expansion of cd71+ erythroid progenitor cells for cell therapy | |
JP2024525910A (en) | Methods for generating populations of immune cells from pluripotent stem cells | |
CN117683713A (en) | In vitro preparation of T lymphocytes and NK cells using human pluripotent stem cells |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 22856818 Country of ref document: EP Kind code of ref document: A1 |
|
WWE | Wipo information: entry into national phase |
Ref document number: 202280055442.9 Country of ref document: CN |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
122 | Ep: pct application non-entry in european phase |
Ref document number: 22856818 Country of ref document: EP Kind code of ref document: A1 |