WO2013188427A1 - Improved methods of cell culture for adoptive cell therapy - Google Patents
Improved methods of cell culture for adoptive cell therapy Download PDFInfo
- Publication number
- WO2013188427A1 WO2013188427A1 PCT/US2013/045209 US2013045209W WO2013188427A1 WO 2013188427 A1 WO2013188427 A1 WO 2013188427A1 US 2013045209 W US2013045209 W US 2013045209W WO 2013188427 A1 WO2013188427 A1 WO 2013188427A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- cells
- cell
- antigen
- vehicles
- population
- Prior art date
Links
- 238000000034 method Methods 0.000 title claims description 116
- 238000004113 cell culture Methods 0.000 title claims description 19
- 238000011467 adoptive cell therapy Methods 0.000 title abstract description 15
- 230000001976 improved effect Effects 0.000 title description 10
- 210000004027 cell Anatomy 0.000 claims abstract description 459
- 239000000427 antigen Substances 0.000 claims abstract description 222
- 108091007433 antigens Proteins 0.000 claims abstract description 222
- 102000036639 antigens Human genes 0.000 claims abstract description 222
- 210000001744 T-lymphocyte Anatomy 0.000 claims abstract description 182
- 230000012010 growth Effects 0.000 claims description 110
- 210000003819 peripheral blood mononuclear cell Anatomy 0.000 claims description 35
- 239000000463 material Substances 0.000 claims description 28
- 230000008569 process Effects 0.000 claims description 20
- 210000004700 fetal blood Anatomy 0.000 claims description 15
- 230000035755 proliferation Effects 0.000 claims description 12
- 241000700605 Viruses Species 0.000 claims description 11
- 230000004936 stimulating effect Effects 0.000 claims description 9
- 210000005260 human cell Anatomy 0.000 claims description 8
- 241000894006 Bacteria Species 0.000 claims description 4
- 241000233866 Fungi Species 0.000 claims description 4
- 241000251468 Actinopterygii Species 0.000 claims description 3
- 241000938605 Crocodylia Species 0.000 claims description 3
- 241000243142 Porifera Species 0.000 claims description 3
- 244000045947 parasite Species 0.000 claims description 3
- 230000004044 response Effects 0.000 claims description 2
- 238000004519 manufacturing process Methods 0.000 abstract description 97
- 230000001225 therapeutic effect Effects 0.000 abstract description 92
- 230000008901 benefit Effects 0.000 abstract description 34
- 108010083359 Antigen Receptors Proteins 0.000 abstract description 23
- 102000006306 Antigen Receptors Human genes 0.000 abstract description 23
- 208000024908 graft versus host disease Diseases 0.000 abstract description 18
- 238000001727 in vivo Methods 0.000 abstract description 14
- 230000000977 initiatory effect Effects 0.000 abstract description 11
- 230000000735 allogeneic effect Effects 0.000 abstract description 7
- 239000002609 medium Substances 0.000 description 65
- 238000007796 conventional method Methods 0.000 description 53
- 210000000612 antigen-presenting cell Anatomy 0.000 description 35
- 239000007789 gas Substances 0.000 description 35
- 239000000203 mixture Substances 0.000 description 35
- 206010028980 Neoplasm Diseases 0.000 description 30
- 102000004127 Cytokines Human genes 0.000 description 24
- 108090000695 Cytokines Proteins 0.000 description 24
- 230000001965 increasing effect Effects 0.000 description 21
- 210000004881 tumor cell Anatomy 0.000 description 20
- 238000012360 testing method Methods 0.000 description 19
- 230000000638 stimulation Effects 0.000 description 17
- 208000009329 Graft vs Host Disease Diseases 0.000 description 16
- 241000701044 Human gammaherpesvirus 4 Species 0.000 description 16
- 102000004388 Interleukin-4 Human genes 0.000 description 16
- 108090000978 Interleukin-4 Proteins 0.000 description 16
- 108010008281 Recombinant Fusion Proteins Proteins 0.000 description 16
- 102000007056 Recombinant Fusion Proteins Human genes 0.000 description 16
- 108090000765 processed proteins & peptides Proteins 0.000 description 16
- 108010002586 Interleukin-7 Proteins 0.000 description 15
- 238000002659 cell therapy Methods 0.000 description 14
- 108090000623 proteins and genes Proteins 0.000 description 13
- 101710120463 Prostate stem cell antigen Proteins 0.000 description 12
- 102100036735 Prostate stem cell antigen Human genes 0.000 description 12
- 230000002829 reductive effect Effects 0.000 description 12
- 238000011282 treatment Methods 0.000 description 12
- 239000002246 antineoplastic agent Substances 0.000 description 11
- 238000013459 approach Methods 0.000 description 10
- 230000010261 cell growth Effects 0.000 description 10
- 229940127089 cytotoxic agent Drugs 0.000 description 10
- 230000014509 gene expression Effects 0.000 description 10
- 108010019670 Chimeric Antigen Receptors Proteins 0.000 description 9
- 206010061218 Inflammation Diseases 0.000 description 9
- 108010002350 Interleukin-2 Proteins 0.000 description 9
- 102000000588 Interleukin-2 Human genes 0.000 description 9
- 238000012258 culturing Methods 0.000 description 9
- 230000004054 inflammatory process Effects 0.000 description 9
- 238000012239 gene modification Methods 0.000 description 8
- 230000005017 genetic modification Effects 0.000 description 8
- 235000013617 genetically modified food Nutrition 0.000 description 8
- 210000000987 immune system Anatomy 0.000 description 8
- 230000000670 limiting effect Effects 0.000 description 8
- 108091008874 T cell receptors Proteins 0.000 description 7
- 102000016266 T-Cell Antigen Receptors Human genes 0.000 description 7
- 238000004458 analytical method Methods 0.000 description 7
- 238000003556 assay Methods 0.000 description 7
- 239000012737 fresh medium Substances 0.000 description 7
- 230000006870 function Effects 0.000 description 7
- 230000028993 immune response Effects 0.000 description 7
- 230000002147 killing effect Effects 0.000 description 7
- 230000002062 proliferating effect Effects 0.000 description 7
- 238000010186 staining Methods 0.000 description 7
- -1 succinimidyl ester Chemical class 0.000 description 7
- ZCYVEMRRCGMTRW-UHFFFAOYSA-N 7553-56-2 Chemical compound [I] ZCYVEMRRCGMTRW-UHFFFAOYSA-N 0.000 description 6
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 6
- 230000000840 anti-viral effect Effects 0.000 description 6
- 210000003719 b-lymphocyte Anatomy 0.000 description 6
- 201000011510 cancer Diseases 0.000 description 6
- 238000002512 chemotherapy Methods 0.000 description 6
- 230000003247 decreasing effect Effects 0.000 description 6
- 230000000694 effects Effects 0.000 description 6
- 238000009169 immunotherapy Methods 0.000 description 6
- 229910052740 iodine Inorganic materials 0.000 description 6
- 239000011630 iodine Substances 0.000 description 6
- 231100000419 toxicity Toxicity 0.000 description 6
- 230000001988 toxicity Effects 0.000 description 6
- 238000012384 transportation and delivery Methods 0.000 description 6
- 230000003442 weekly effect Effects 0.000 description 6
- 208000023275 Autoimmune disease Diseases 0.000 description 5
- 102100022005 B-lymphocyte antigen CD20 Human genes 0.000 description 5
- 241000701022 Cytomegalovirus Species 0.000 description 5
- 238000002965 ELISA Methods 0.000 description 5
- 101000897405 Homo sapiens B-lymphocyte antigen CD20 Proteins 0.000 description 5
- 241000699670 Mus sp. Species 0.000 description 5
- 101000588258 Taenia solium Paramyosin Proteins 0.000 description 5
- 238000010790 dilution Methods 0.000 description 5
- 239000012895 dilution Substances 0.000 description 5
- LOKCTEFSRHRXRJ-UHFFFAOYSA-I dipotassium trisodium dihydrogen phosphate hydrogen phosphate dichloride Chemical compound P(=O)(O)(O)[O-].[K+].P(=O)(O)([O-])[O-].[Na+].[Na+].[Cl-].[K+].[Cl-].[Na+] LOKCTEFSRHRXRJ-UHFFFAOYSA-I 0.000 description 5
- 238000002474 experimental method Methods 0.000 description 5
- 238000005206 flow analysis Methods 0.000 description 5
- 239000002953 phosphate buffered saline Substances 0.000 description 5
- 230000009467 reduction Effects 0.000 description 5
- 230000002459 sustained effect Effects 0.000 description 5
- 210000001519 tissue Anatomy 0.000 description 5
- 102100021592 Interleukin-7 Human genes 0.000 description 4
- 239000002671 adjuvant Substances 0.000 description 4
- 230000000259 anti-tumor effect Effects 0.000 description 4
- 230000008859 change Effects 0.000 description 4
- 239000003814 drug Substances 0.000 description 4
- 230000008030 elimination Effects 0.000 description 4
- 238000003379 elimination reaction Methods 0.000 description 4
- 239000012091 fetal bovine serum Substances 0.000 description 4
- 238000000684 flow cytometry Methods 0.000 description 4
- 238000003384 imaging method Methods 0.000 description 4
- 238000010348 incorporation Methods 0.000 description 4
- 238000001802 infusion Methods 0.000 description 4
- 210000004962 mammalian cell Anatomy 0.000 description 4
- 201000001441 melanoma Diseases 0.000 description 4
- 238000010899 nucleation Methods 0.000 description 4
- 230000002688 persistence Effects 0.000 description 4
- 102000005962 receptors Human genes 0.000 description 4
- 108020003175 receptors Proteins 0.000 description 4
- 238000012546 transfer Methods 0.000 description 4
- 210000003171 tumor-infiltrating lymphocyte Anatomy 0.000 description 4
- 241000701161 unidentified adenovirus Species 0.000 description 4
- 241001430294 unidentified retrovirus Species 0.000 description 4
- 239000013598 vector Substances 0.000 description 4
- 230000035899 viability Effects 0.000 description 4
- 230000003612 virological effect Effects 0.000 description 4
- VYZAMTAEIAYCRO-UHFFFAOYSA-N Chromium Chemical compound [Cr] VYZAMTAEIAYCRO-UHFFFAOYSA-N 0.000 description 3
- WQZGKKKJIJFFOK-GASJEMHNSA-N Glucose Natural products OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 3
- 101001136981 Homo sapiens Proteasome subunit beta type-9 Proteins 0.000 description 3
- 101000914484 Homo sapiens T-lymphocyte activation antigen CD80 Proteins 0.000 description 3
- 241000713666 Lentivirus Species 0.000 description 3
- 108700018351 Major Histocompatibility Complex Proteins 0.000 description 3
- 108010004729 Phycoerythrin Proteins 0.000 description 3
- 102100035764 Proteasome subunit beta type-9 Human genes 0.000 description 3
- 238000001190 Q-PCR Methods 0.000 description 3
- 239000012980 RPMI-1640 medium Substances 0.000 description 3
- 241000725643 Respiratory syncytial virus Species 0.000 description 3
- 241000700584 Simplexvirus Species 0.000 description 3
- 102100027222 T-lymphocyte activation antigen CD80 Human genes 0.000 description 3
- 108700019146 Transgenes Proteins 0.000 description 3
- 239000004599 antimicrobial Substances 0.000 description 3
- 230000032823 cell division Effects 0.000 description 3
- 230000003833 cell viability Effects 0.000 description 3
- 239000011651 chromium Substances 0.000 description 3
- 229910052804 chromium Inorganic materials 0.000 description 3
- 150000001875 compounds Chemical class 0.000 description 3
- 230000001186 cumulative effect Effects 0.000 description 3
- 230000016396 cytokine production Effects 0.000 description 3
- 102000003675 cytokine receptors Human genes 0.000 description 3
- 108010057085 cytokine receptors Proteins 0.000 description 3
- 230000003467 diminishing effect Effects 0.000 description 3
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 3
- 238000003114 enzyme-linked immunosorbent spot assay Methods 0.000 description 3
- 230000001747 exhibiting effect Effects 0.000 description 3
- 238000000338 in vitro Methods 0.000 description 3
- 238000011534 incubation Methods 0.000 description 3
- 210000003734 kidney Anatomy 0.000 description 3
- 238000002372 labelling Methods 0.000 description 3
- 210000004185 liver Anatomy 0.000 description 3
- 230000005012 migration Effects 0.000 description 3
- 238000013508 migration Methods 0.000 description 3
- 210000000822 natural killer cell Anatomy 0.000 description 3
- 235000015097 nutrients Nutrition 0.000 description 3
- 210000000056 organ Anatomy 0.000 description 3
- 210000005259 peripheral blood Anatomy 0.000 description 3
- 239000011886 peripheral blood Substances 0.000 description 3
- 102000004169 proteins and genes Human genes 0.000 description 3
- 210000003289 regulatory T cell Anatomy 0.000 description 3
- 230000000284 resting effect Effects 0.000 description 3
- 230000003068 static effect Effects 0.000 description 3
- 239000000126 substance Substances 0.000 description 3
- 230000020382 suppression by virus of host antigen processing and presentation of peptide antigen via MHC class I Effects 0.000 description 3
- 238000002560 therapeutic procedure Methods 0.000 description 3
- 230000009261 transgenic effect Effects 0.000 description 3
- YBJHBAHKTGYVGT-ZKWXMUAHSA-N (+)-Biotin Chemical group N1C(=O)N[C@@H]2[C@H](CCCCC(=O)O)SC[C@@H]21 YBJHBAHKTGYVGT-ZKWXMUAHSA-N 0.000 description 2
- HGINCPLSRVDWNT-UHFFFAOYSA-N Acrolein Chemical compound C=CC=O HGINCPLSRVDWNT-UHFFFAOYSA-N 0.000 description 2
- 108700028369 Alleles Proteins 0.000 description 2
- 201000004384 Alopecia Diseases 0.000 description 2
- 102100022718 Atypical chemokine receptor 2 Human genes 0.000 description 2
- 102100027207 CD27 antigen Human genes 0.000 description 2
- 101710098119 Chaperonin GroEL 2 Proteins 0.000 description 2
- 241000724252 Cucumber mosaic virus Species 0.000 description 2
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N Dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 2
- 102000004190 Enzymes Human genes 0.000 description 2
- 108090000790 Enzymes Proteins 0.000 description 2
- 108091034120 Epstein–Barr virus-encoded small RNA Proteins 0.000 description 2
- 108010017213 Granulocyte-Macrophage Colony-Stimulating Factor Proteins 0.000 description 2
- 241000696272 Gull adenovirus Species 0.000 description 2
- 108010033040 Histones Proteins 0.000 description 2
- 101000678892 Homo sapiens Atypical chemokine receptor 2 Proteins 0.000 description 2
- 101000914511 Homo sapiens CD27 antigen Proteins 0.000 description 2
- 101001057504 Homo sapiens Interferon-stimulated gene 20 kDa protein Proteins 0.000 description 2
- 101001055144 Homo sapiens Interleukin-2 receptor subunit alpha Proteins 0.000 description 2
- 101001033312 Homo sapiens Interleukin-4 receptor subunit alpha Proteins 0.000 description 2
- 101001018097 Homo sapiens L-selectin Proteins 0.000 description 2
- 101000914514 Homo sapiens T-cell-specific surface glycoprotein CD28 Proteins 0.000 description 2
- 241000701085 Human alphaherpesvirus 3 Species 0.000 description 2
- 241000701041 Human betaherpesvirus 7 Species 0.000 description 2
- 241000701027 Human herpesvirus 6 Species 0.000 description 2
- MHAJPDPJQMAIIY-UHFFFAOYSA-N Hydrogen peroxide Chemical compound OO MHAJPDPJQMAIIY-UHFFFAOYSA-N 0.000 description 2
- 102000026633 IL6 Human genes 0.000 description 2
- 102000003814 Interleukin-10 Human genes 0.000 description 2
- 108090000174 Interleukin-10 Proteins 0.000 description 2
- 108010065805 Interleukin-12 Proteins 0.000 description 2
- 102000013462 Interleukin-12 Human genes 0.000 description 2
- 108090000176 Interleukin-13 Proteins 0.000 description 2
- 102000003816 Interleukin-13 Human genes 0.000 description 2
- 108090000172 Interleukin-15 Proteins 0.000 description 2
- 102000003812 Interleukin-15 Human genes 0.000 description 2
- 102100026878 Interleukin-2 receptor subunit alpha Human genes 0.000 description 2
- 102100030704 Interleukin-21 Human genes 0.000 description 2
- 108090001005 Interleukin-6 Proteins 0.000 description 2
- 108010036012 Iodide peroxidase Proteins 0.000 description 2
- ZDXPYRJPNDTMRX-VKHMYHEASA-N L-glutamine Chemical compound OC(=O)[C@@H](N)CCC(N)=O ZDXPYRJPNDTMRX-VKHMYHEASA-N 0.000 description 2
- 229930182816 L-glutamine Natural products 0.000 description 2
- 102100033467 L-selectin Human genes 0.000 description 2
- 206010028116 Mucosal inflammation Diseases 0.000 description 2
- 201000010927 Mucositis Diseases 0.000 description 2
- 108010042215 OX40 Ligand Proteins 0.000 description 2
- 208000002606 Paramyxoviridae Infections Diseases 0.000 description 2
- 239000012979 RPMI medium Substances 0.000 description 2
- 102100027213 T-cell-specific surface glycoprotein CD28 Human genes 0.000 description 2
- 102000006601 Thymidine Kinase Human genes 0.000 description 2
- 108020004440 Thymidine kinase Proteins 0.000 description 2
- 208000024770 Thyroid neoplasm Diseases 0.000 description 2
- 102100026890 Tumor necrosis factor ligand superfamily member 4 Human genes 0.000 description 2
- 208000036142 Viral infection Diseases 0.000 description 2
- 238000009825 accumulation Methods 0.000 description 2
- 230000004913 activation Effects 0.000 description 2
- 230000006786 activation induced cell death Effects 0.000 description 2
- 230000003698 anagen phase Effects 0.000 description 2
- 230000001640 apoptogenic effect Effects 0.000 description 2
- 230000003190 augmentative effect Effects 0.000 description 2
- 230000009286 beneficial effect Effects 0.000 description 2
- 229960002685 biotin Drugs 0.000 description 2
- 239000011616 biotin Substances 0.000 description 2
- 230000030833 cell death Effects 0.000 description 2
- 239000002771 cell marker Substances 0.000 description 2
- 239000006285 cell suspension Substances 0.000 description 2
- 239000003795 chemical substances by application Substances 0.000 description 2
- 229940044683 chemotherapy drug Drugs 0.000 description 2
- 238000011109 contamination Methods 0.000 description 2
- 238000012136 culture method Methods 0.000 description 2
- 230000001461 cytolytic effect Effects 0.000 description 2
- 230000006378 damage Effects 0.000 description 2
- 230000007423 decrease Effects 0.000 description 2
- 238000001514 detection method Methods 0.000 description 2
- 238000009826 distribution Methods 0.000 description 2
- 229940079593 drug Drugs 0.000 description 2
- 238000011156 evaluation Methods 0.000 description 2
- 239000012894 fetal calf serum Substances 0.000 description 2
- MHMNJMPURVTYEJ-UHFFFAOYSA-N fluorescein-5-isothiocyanate Chemical compound O1C(=O)C2=CC(N=C=S)=CC=C2C21C1=CC=C(O)C=C1OC1=CC(O)=CC=C21 MHMNJMPURVTYEJ-UHFFFAOYSA-N 0.000 description 2
- IRSCQMHQWWYFCW-UHFFFAOYSA-N ganciclovir Chemical compound O=C1NC(N)=NC2=C1N=CN2COC(CO)CO IRSCQMHQWWYFCW-UHFFFAOYSA-N 0.000 description 2
- 229960002963 ganciclovir Drugs 0.000 description 2
- 210000001035 gastrointestinal tract Anatomy 0.000 description 2
- 239000008103 glucose Substances 0.000 description 2
- 230000006028 immune-suppresssive effect Effects 0.000 description 2
- 238000013394 immunophenotyping Methods 0.000 description 2
- 230000001506 immunosuppresive effect Effects 0.000 description 2
- 238000011503 in vivo imaging Methods 0.000 description 2
- 230000006698 induction Effects 0.000 description 2
- 230000001939 inductive effect Effects 0.000 description 2
- 230000003993 interaction Effects 0.000 description 2
- 108010074108 interleukin-21 Proteins 0.000 description 2
- 208000032839 leukemia Diseases 0.000 description 2
- 239000003446 ligand Substances 0.000 description 2
- 239000012528 membrane Substances 0.000 description 2
- 230000004048 modification Effects 0.000 description 2
- 238000012986 modification Methods 0.000 description 2
- 230000001338 necrotic effect Effects 0.000 description 2
- 238000009206 nuclear medicine Methods 0.000 description 2
- 230000009437 off-target effect Effects 0.000 description 2
- 230000036961 partial effect Effects 0.000 description 2
- 239000013612 plasmid Substances 0.000 description 2
- 102000004196 processed proteins & peptides Human genes 0.000 description 2
- 239000000700 radioactive tracer Substances 0.000 description 2
- 230000001177 retroviral effect Effects 0.000 description 2
- 229960004641 rituximab Drugs 0.000 description 2
- 150000003384 small molecules Chemical class 0.000 description 2
- 230000002269 spontaneous effect Effects 0.000 description 2
- 238000007619 statistical method 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
- 239000000758 substrate Substances 0.000 description 2
- 238000007910 systemic administration Methods 0.000 description 2
- 230000009885 systemic effect Effects 0.000 description 2
- 238000001890 transfection Methods 0.000 description 2
- 230000009385 viral infection Effects 0.000 description 2
- 239000013603 viral vector Substances 0.000 description 2
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 1
- LKKMLIBUAXYLOY-UHFFFAOYSA-N 3-Amino-1-methyl-5H-pyrido[4,3-b]indole Chemical compound N1C2=CC=CC=C2C2=C1C=C(N)N=C2C LKKMLIBUAXYLOY-UHFFFAOYSA-N 0.000 description 1
- WEVYNIUIFUYDGI-UHFFFAOYSA-N 3-[6-[4-(trifluoromethoxy)anilino]-4-pyrimidinyl]benzamide Chemical compound NC(=O)C1=CC=CC(C=2N=CN=C(NC=3C=CC(OC(F)(F)F)=CC=3)C=2)=C1 WEVYNIUIFUYDGI-UHFFFAOYSA-N 0.000 description 1
- BZTDTCNHAFUJOG-UHFFFAOYSA-N 6-carboxyfluorescein Chemical compound C12=CC=C(O)C=C2OC2=CC(O)=CC=C2C11OC(=O)C2=CC=C(C(=O)O)C=C21 BZTDTCNHAFUJOG-UHFFFAOYSA-N 0.000 description 1
- YXHLJMWYDTXDHS-IRFLANFNSA-N 7-aminoactinomycin D Chemical compound C[C@H]1OC(=O)[C@H](C(C)C)N(C)C(=O)CN(C)C(=O)[C@@H]2CCCN2C(=O)[C@@H](C(C)C)NC(=O)[C@H]1NC(=O)C1=C(N)C(=O)C(C)=C2OC(C(C)=C(N)C=C3C(=O)N[C@@H]4C(=O)N[C@@H](C(N5CCC[C@H]5C(=O)N(C)CC(=O)N(C)[C@@H](C(C)C)C(=O)O[C@@H]4C)=O)C(C)C)=C3N=C21 YXHLJMWYDTXDHS-IRFLANFNSA-N 0.000 description 1
- 108700012813 7-aminoactinomycin D Proteins 0.000 description 1
- 241000004176 Alphacoronavirus Species 0.000 description 1
- 102000000412 Annexin Human genes 0.000 description 1
- 108050008874 Annexin Proteins 0.000 description 1
- 108091023037 Aptamer Proteins 0.000 description 1
- 241000238421 Arthropoda Species 0.000 description 1
- 208000003950 B-cell lymphoma Diseases 0.000 description 1
- 101150009389 BZLF1 gene Proteins 0.000 description 1
- 102100021663 Baculoviral IAP repeat-containing protein 5 Human genes 0.000 description 1
- 241000124740 Bocaparvovirus Species 0.000 description 1
- 206010065553 Bone marrow failure Diseases 0.000 description 1
- 102100031172 C-C chemokine receptor type 1 Human genes 0.000 description 1
- 101710149814 C-C chemokine receptor type 1 Proteins 0.000 description 1
- 102100031151 C-C chemokine receptor type 2 Human genes 0.000 description 1
- 101710149815 C-C chemokine receptor type 2 Proteins 0.000 description 1
- 102100024167 C-C chemokine receptor type 3 Human genes 0.000 description 1
- 101710149862 C-C chemokine receptor type 3 Proteins 0.000 description 1
- 102100037853 C-C chemokine receptor type 4 Human genes 0.000 description 1
- 101710149863 C-C chemokine receptor type 4 Proteins 0.000 description 1
- 102100035875 C-C chemokine receptor type 5 Human genes 0.000 description 1
- 101710149870 C-C chemokine receptor type 5 Proteins 0.000 description 1
- 102100036301 C-C chemokine receptor type 7 Human genes 0.000 description 1
- 102100036305 C-C chemokine receptor type 8 Human genes 0.000 description 1
- 102100025074 C-C chemokine receptor-like 2 Human genes 0.000 description 1
- 102100036166 C-X-C chemokine receptor type 1 Human genes 0.000 description 1
- 102100028989 C-X-C chemokine receptor type 2 Human genes 0.000 description 1
- 102100031658 C-X-C chemokine receptor type 5 Human genes 0.000 description 1
- 102100025618 C-X-C chemokine receptor type 6 Human genes 0.000 description 1
- 102100032912 CD44 antigen Human genes 0.000 description 1
- 102000008203 CTLA-4 Antigen Human genes 0.000 description 1
- 108010021064 CTLA-4 Antigen Proteins 0.000 description 1
- 229940045513 CTLA4 antagonist Drugs 0.000 description 1
- 102100025570 Cancer/testis antigen 1 Human genes 0.000 description 1
- 102000014914 Carrier Proteins Human genes 0.000 description 1
- 102000004039 Caspase-9 Human genes 0.000 description 1
- 108090000566 Caspase-9 Proteins 0.000 description 1
- 102000000844 Cell Surface Receptors Human genes 0.000 description 1
- 108010001857 Cell Surface Receptors Proteins 0.000 description 1
- 102100025064 Cellular tumor antigen p53 Human genes 0.000 description 1
- 102100035294 Chemokine XC receptor 1 Human genes 0.000 description 1
- 102000009410 Chemokine receptor Human genes 0.000 description 1
- 108050000299 Chemokine receptor Proteins 0.000 description 1
- 102000019034 Chemokines Human genes 0.000 description 1
- 108010012236 Chemokines Proteins 0.000 description 1
- 241001227713 Chiron Species 0.000 description 1
- 102100038449 Claudin-6 Human genes 0.000 description 1
- 108090000229 Claudin-6 Proteins 0.000 description 1
- 206010010144 Completed suicide Diseases 0.000 description 1
- 241000700108 Ctenophora <comb jellyfish phylum> Species 0.000 description 1
- 101100239628 Danio rerio myca gene Proteins 0.000 description 1
- 206010011968 Decreased immune responsiveness Diseases 0.000 description 1
- 241000702421 Dependoparvovirus Species 0.000 description 1
- 206010012735 Diarrhoea Diseases 0.000 description 1
- 101150059079 EBNA1 gene Proteins 0.000 description 1
- 101150029707 ERBB2 gene Proteins 0.000 description 1
- 241000258955 Echinodermata Species 0.000 description 1
- 241000132179 Eurotium medium Species 0.000 description 1
- 102000010579 Fas-Associated Death Domain Protein Human genes 0.000 description 1
- 108010077716 Fas-Associated Death Domain Protein Proteins 0.000 description 1
- 102100041003 Glutamate carboxypeptidase 2 Human genes 0.000 description 1
- 102000004457 Granulocyte-Macrophage Colony-Stimulating Factor Human genes 0.000 description 1
- 102100039620 Granulocyte-macrophage colony-stimulating factor Human genes 0.000 description 1
- 108010074032 HLA-A2 Antigen Proteins 0.000 description 1
- 102000025850 HLA-A2 Antigen Human genes 0.000 description 1
- 108010034115 HLA-A29 antigen Proteins 0.000 description 1
- 208000002250 Hematologic Neoplasms Diseases 0.000 description 1
- 102100034523 Histone H4 Human genes 0.000 description 1
- 208000017604 Hodgkin disease Diseases 0.000 description 1
- 208000021519 Hodgkin lymphoma Diseases 0.000 description 1
- 208000010747 Hodgkins lymphoma Diseases 0.000 description 1
- 101000777558 Homo sapiens C-C chemokine receptor type 10 Proteins 0.000 description 1
- 101000716068 Homo sapiens C-C chemokine receptor type 6 Proteins 0.000 description 1
- 101000716065 Homo sapiens C-C chemokine receptor type 7 Proteins 0.000 description 1
- 101000716063 Homo sapiens C-C chemokine receptor type 8 Proteins 0.000 description 1
- 101000716070 Homo sapiens C-C chemokine receptor type 9 Proteins 0.000 description 1
- 101000947174 Homo sapiens C-X-C chemokine receptor type 1 Proteins 0.000 description 1
- 101000922405 Homo sapiens C-X-C chemokine receptor type 5 Proteins 0.000 description 1
- 101000856683 Homo sapiens C-X-C chemokine receptor type 6 Proteins 0.000 description 1
- 101000868273 Homo sapiens CD44 antigen Proteins 0.000 description 1
- 101000856237 Homo sapiens Cancer/testis antigen 1 Proteins 0.000 description 1
- 101000914324 Homo sapiens Carcinoembryonic antigen-related cell adhesion molecule 5 Proteins 0.000 description 1
- 101000914321 Homo sapiens Carcinoembryonic antigen-related cell adhesion molecule 7 Proteins 0.000 description 1
- 101000721661 Homo sapiens Cellular tumor antigen p53 Proteins 0.000 description 1
- 101000804783 Homo sapiens Chemokine XC receptor 1 Proteins 0.000 description 1
- 101000892862 Homo sapiens Glutamate carboxypeptidase 2 Proteins 0.000 description 1
- 101001002657 Homo sapiens Interleukin-2 Proteins 0.000 description 1
- 101000853000 Homo sapiens Interleukin-26 Proteins 0.000 description 1
- 101100495232 Homo sapiens MS4A1 gene Proteins 0.000 description 1
- 101000578784 Homo sapiens Melanoma antigen recognized by T-cells 1 Proteins 0.000 description 1
- 101000581981 Homo sapiens Neural cell adhesion molecule 1 Proteins 0.000 description 1
- 101000617725 Homo sapiens Pregnancy-specific beta-1-glycoprotein 2 Proteins 0.000 description 1
- 101000880770 Homo sapiens Protein SSX2 Proteins 0.000 description 1
- 101000880774 Homo sapiens Protein SSX4 Proteins 0.000 description 1
- 101000635938 Homo sapiens Transforming growth factor beta-1 proprotein Proteins 0.000 description 1
- 108010001336 Horseradish Peroxidase Proteins 0.000 description 1
- 241000702617 Human parvovirus B19 Species 0.000 description 1
- 241000829111 Human polyomavirus 1 Species 0.000 description 1
- 206010062016 Immunosuppression Diseases 0.000 description 1
- 102000003810 Interleukin-18 Human genes 0.000 description 1
- 108090000171 Interleukin-18 Proteins 0.000 description 1
- 102100036679 Interleukin-26 Human genes 0.000 description 1
- 108010066979 Interleukin-27 Proteins 0.000 description 1
- 102100036678 Interleukin-27 subunit alpha Human genes 0.000 description 1
- 108010018951 Interleukin-8B Receptors Proteins 0.000 description 1
- ZCYVEMRRCGMTRW-AHCXROLUSA-N Iodine-123 Chemical compound [123I] ZCYVEMRRCGMTRW-AHCXROLUSA-N 0.000 description 1
- 241000701460 JC polyomavirus Species 0.000 description 1
- 101150113776 LMP1 gene Proteins 0.000 description 1
- 206010025323 Lymphomas Diseases 0.000 description 1
- 101150039798 MYC gene Proteins 0.000 description 1
- 102000005727 Mammaglobin A Human genes 0.000 description 1
- 108010031030 Mammaglobin A Proteins 0.000 description 1
- 108010076502 Matrix Metalloproteinase 11 Proteins 0.000 description 1
- 102100028389 Melanoma antigen recognized by T-cells 1 Human genes 0.000 description 1
- 206010027476 Metastases Diseases 0.000 description 1
- 241001465754 Metazoa Species 0.000 description 1
- 241000237852 Mollusca Species 0.000 description 1
- 208000002454 Nasopharyngeal Carcinoma Diseases 0.000 description 1
- 206010061306 Nasopharyngeal cancer Diseases 0.000 description 1
- 206010028813 Nausea Diseases 0.000 description 1
- 102100027347 Neural cell adhesion molecule 1 Human genes 0.000 description 1
- 206010030113 Oedema Diseases 0.000 description 1
- 240000007019 Oxalis corniculata Species 0.000 description 1
- 102000036673 PRAME Human genes 0.000 description 1
- 108060006580 PRAME Proteins 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
- 102000035195 Peptidases Human genes 0.000 description 1
- 108091005804 Peptidases Proteins 0.000 description 1
- 108010067902 Peptide Library Proteins 0.000 description 1
- 102100021768 Phosphoserine aminotransferase Human genes 0.000 description 1
- 108091000080 Phosphotransferase Proteins 0.000 description 1
- 206010035226 Plasma cell myeloma Diseases 0.000 description 1
- 102100022019 Pregnancy-specific beta-1-glycoprotein 2 Human genes 0.000 description 1
- 108010072866 Prostate-Specific Antigen Proteins 0.000 description 1
- 102100035703 Prostatic acid phosphatase Human genes 0.000 description 1
- 102100037686 Protein SSX2 Human genes 0.000 description 1
- 102100037727 Protein SSX4 Human genes 0.000 description 1
- 101100247004 Rattus norvegicus Qsox1 gene Proteins 0.000 description 1
- 206010038687 Respiratory distress Diseases 0.000 description 1
- 241000702670 Rotavirus Species 0.000 description 1
- 102000017303 Stromelysin-3 Human genes 0.000 description 1
- 238000000692 Student's t-test Methods 0.000 description 1
- 108010002687 Survivin Proteins 0.000 description 1
- 101150003725 TK gene Proteins 0.000 description 1
- 108700012920 TNF Proteins 0.000 description 1
- 208000024799 Thyroid disease Diseases 0.000 description 1
- 102000014267 Thyroid peroxidases Human genes 0.000 description 1
- 102000009618 Transforming Growth Factors Human genes 0.000 description 1
- 108010009583 Transforming Growth Factors Proteins 0.000 description 1
- 102100030742 Transforming growth factor beta-1 proprotein Human genes 0.000 description 1
- 102000008579 Transposases Human genes 0.000 description 1
- 108010020764 Transposases Proteins 0.000 description 1
- 102100022153 Tumor necrosis factor receptor superfamily member 4 Human genes 0.000 description 1
- 101710165473 Tumor necrosis factor receptor superfamily member 4 Proteins 0.000 description 1
- 102100039094 Tyrosinase Human genes 0.000 description 1
- 108060008724 Tyrosinase Proteins 0.000 description 1
- 206010047700 Vomiting Diseases 0.000 description 1
- 108700020467 WT1 Proteins 0.000 description 1
- 101150084041 WT1 gene Proteins 0.000 description 1
- 101100459258 Xenopus laevis myc-a gene Proteins 0.000 description 1
- 239000008186 active pharmaceutical agent Substances 0.000 description 1
- 230000001154 acute effect Effects 0.000 description 1
- 239000000654 additive Substances 0.000 description 1
- 230000000996 additive effect Effects 0.000 description 1
- 230000001464 adherent effect Effects 0.000 description 1
- 239000000556 agonist Substances 0.000 description 1
- 108700025316 aldesleukin Proteins 0.000 description 1
- 108010004469 allophycocyanin Proteins 0.000 description 1
- 231100000360 alopecia Toxicity 0.000 description 1
- 102000013529 alpha-Fetoproteins Human genes 0.000 description 1
- 108010026331 alpha-Fetoproteins Proteins 0.000 description 1
- 239000005557 antagonist Substances 0.000 description 1
- 230000000843 anti-fungal effect Effects 0.000 description 1
- 230000000845 anti-microbial effect Effects 0.000 description 1
- 230000005809 anti-tumor immunity Effects 0.000 description 1
- 230000000890 antigenic effect Effects 0.000 description 1
- 229940041181 antineoplastic drug Drugs 0.000 description 1
- 239000003096 antiparasitic agent Substances 0.000 description 1
- 229940125687 antiparasitic agent Drugs 0.000 description 1
- 230000006907 apoptotic process Effects 0.000 description 1
- 239000012298 atmosphere Substances 0.000 description 1
- QVGXLLKOCUKJST-UHFFFAOYSA-N atomic oxygen Chemical compound [O] QVGXLLKOCUKJST-UHFFFAOYSA-N 0.000 description 1
- 230000001363 autoimmune Effects 0.000 description 1
- 239000012148 binding buffer Substances 0.000 description 1
- 108091008324 binding proteins Proteins 0.000 description 1
- 230000015572 biosynthetic process Effects 0.000 description 1
- 235000020958 biotin Nutrition 0.000 description 1
- 210000004369 blood Anatomy 0.000 description 1
- 239000008280 blood Substances 0.000 description 1
- 210000000601 blood cell Anatomy 0.000 description 1
- 210000001185 bone marrow Anatomy 0.000 description 1
- 210000004556 brain Anatomy 0.000 description 1
- 210000000481 breast Anatomy 0.000 description 1
- 239000000969 carrier Substances 0.000 description 1
- 238000010370 cell cloning Methods 0.000 description 1
- 239000013592 cell lysate Substances 0.000 description 1
- 238000002737 cell proliferation kit Methods 0.000 description 1
- 238000001311 chemical methods and process Methods 0.000 description 1
- 230000003399 chemotactic effect Effects 0.000 description 1
- 239000012829 chemotherapy agent Substances 0.000 description 1
- 229930002875 chlorophyll Natural products 0.000 description 1
- 235000019804 chlorophyll Nutrition 0.000 description 1
- ATNHDLDRLWWWCB-AENOIHSZSA-M chlorophyll a Chemical compound C1([C@@H](C(=O)OC)C(=O)C2=C3C)=C2N2C3=CC(C(CC)=C3C)=[N+]4C3=CC3=C(C=C)C(C)=C5N3[Mg-2]42[N+]2=C1[C@@H](CCC(=O)OC\C=C(/C)CCC[C@H](C)CCC[C@H](C)CCCC(C)C)[C@H](C)C2=C5 ATNHDLDRLWWWCB-AENOIHSZSA-M 0.000 description 1
- 230000001684 chronic effect Effects 0.000 description 1
- 239000007376 cm-medium Substances 0.000 description 1
- 238000004132 cross linking Methods 0.000 description 1
- 230000009089 cytolysis Effects 0.000 description 1
- 210000001151 cytotoxic T lymphocyte Anatomy 0.000 description 1
- 230000001472 cytotoxic effect Effects 0.000 description 1
- 238000002784 cytotoxicity assay Methods 0.000 description 1
- 231100000263 cytotoxicity test Toxicity 0.000 description 1
- 230000034994 death Effects 0.000 description 1
- 230000003111 delayed effect Effects 0.000 description 1
- 210000004443 dendritic cell Anatomy 0.000 description 1
- 230000000779 depleting effect Effects 0.000 description 1
- 238000000151 deposition Methods 0.000 description 1
- 238000006471 dimerization reaction Methods 0.000 description 1
- 230000003292 diminished effect Effects 0.000 description 1
- 201000010099 disease Diseases 0.000 description 1
- 238000004520 electroporation Methods 0.000 description 1
- 238000005516 engineering process Methods 0.000 description 1
- 230000000925 erythroid effect Effects 0.000 description 1
- 125000001495 ethyl group Chemical group [H]C([H])([H])C([H])([H])* 0.000 description 1
- 230000002349 favourable effect Effects 0.000 description 1
- 238000001914 filtration Methods 0.000 description 1
- 108010021843 fluorescent protein 583 Proteins 0.000 description 1
- 239000012634 fragment Substances 0.000 description 1
- 238000010353 genetic engineering Methods 0.000 description 1
- 102000054766 genetic haplotypes Human genes 0.000 description 1
- 108010065889 glycyl-leucyl-cysteinyl-threonyl-leucyl-valyl-alanyl-methionyl-leucine Proteins 0.000 description 1
- 239000003102 growth factor Substances 0.000 description 1
- 210000003780 hair follicle Anatomy 0.000 description 1
- 208000024963 hair loss Diseases 0.000 description 1
- 230000003676 hair loss Effects 0.000 description 1
- 230000007407 health benefit Effects 0.000 description 1
- 210000002216 heart Anatomy 0.000 description 1
- 210000003958 hematopoietic stem cell Anatomy 0.000 description 1
- 230000003054 hormonal effect Effects 0.000 description 1
- 230000036039 immunity Effects 0.000 description 1
- 230000002163 immunogen Effects 0.000 description 1
- 230000001024 immunotherapeutic effect Effects 0.000 description 1
- 230000002779 inactivation Effects 0.000 description 1
- 239000000411 inducer Substances 0.000 description 1
- 208000015181 infectious disease Diseases 0.000 description 1
- 230000036512 infertility Effects 0.000 description 1
- 206010022000 influenza Diseases 0.000 description 1
- 238000002347 injection Methods 0.000 description 1
- 239000007924 injection Substances 0.000 description 1
- 238000002955 isolation Methods 0.000 description 1
- 210000000265 leukocyte Anatomy 0.000 description 1
- 238000001638 lipofection Methods 0.000 description 1
- 230000005976 liver dysfunction Effects 0.000 description 1
- 238000011068 loading method Methods 0.000 description 1
- 210000004072 lung Anatomy 0.000 description 1
- 210000004698 lymphocyte Anatomy 0.000 description 1
- 210000002540 macrophage Anatomy 0.000 description 1
- 230000036210 malignancy 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
- 210000004379 membrane Anatomy 0.000 description 1
- 210000003071 memory t lymphocyte Anatomy 0.000 description 1
- 230000009401 metastasis Effects 0.000 description 1
- 206010061289 metastatic neoplasm Diseases 0.000 description 1
- 238000012737 microarray-based gene expression Methods 0.000 description 1
- 244000005700 microbiome Species 0.000 description 1
- 238000000386 microscopy Methods 0.000 description 1
- 210000001616 monocyte Anatomy 0.000 description 1
- 238000010172 mouse model Methods 0.000 description 1
- 238000012243 multiplex automated genomic engineering Methods 0.000 description 1
- 201000000050 myeloid neoplasm Diseases 0.000 description 1
- 239000002105 nanoparticle Substances 0.000 description 1
- 201000011216 nasopharynx carcinoma Diseases 0.000 description 1
- 230000031942 natural killer cell mediated cytotoxicity Effects 0.000 description 1
- 230000008693 nausea Effects 0.000 description 1
- 239000013642 negative control Substances 0.000 description 1
- 230000001613 neoplastic effect Effects 0.000 description 1
- 231100000252 nontoxic Toxicity 0.000 description 1
- 230000003000 nontoxic effect Effects 0.000 description 1
- 230000002018 overexpression Effects 0.000 description 1
- 239000001301 oxygen Substances 0.000 description 1
- 229910052760 oxygen Inorganic materials 0.000 description 1
- 244000052769 pathogen Species 0.000 description 1
- 230000001717 pathogenic effect Effects 0.000 description 1
- 229940049954 penicillin Drugs 0.000 description 1
- 230000000737 periodic effect Effects 0.000 description 1
- 210000004303 peritoneum Anatomy 0.000 description 1
- 102000020233 phosphotransferase Human genes 0.000 description 1
- 238000000053 physical method Methods 0.000 description 1
- 229920001481 poly(stearyl methacrylate) Polymers 0.000 description 1
- 229920001296 polysiloxane Polymers 0.000 description 1
- 238000002600 positron emission tomography Methods 0.000 description 1
- 239000002243 precursor Substances 0.000 description 1
- 230000000861 pro-apoptotic effect Effects 0.000 description 1
- 229940002612 prodrug Drugs 0.000 description 1
- 239000000651 prodrug Substances 0.000 description 1
- 229940087463 proleukin Drugs 0.000 description 1
- 210000002307 prostate Anatomy 0.000 description 1
- 208000023958 prostate neoplasm Diseases 0.000 description 1
- 108010043671 prostatic acid phosphatase Proteins 0.000 description 1
- 235000019833 protease Nutrition 0.000 description 1
- 238000003908 quality control method Methods 0.000 description 1
- 230000002285 radioactive effect Effects 0.000 description 1
- 102000016914 ras Proteins Human genes 0.000 description 1
- 108010014186 ras Proteins Proteins 0.000 description 1
- 230000009257 reactivity Effects 0.000 description 1
- 230000008085 renal dysfunction Effects 0.000 description 1
- 230000003252 repetitive effect Effects 0.000 description 1
- 238000011160 research Methods 0.000 description 1
- 238000012552 review Methods 0.000 description 1
- 210000002966 serum Anatomy 0.000 description 1
- 238000002603 single-photon emission computed tomography Methods 0.000 description 1
- 230000005783 single-strand break Effects 0.000 description 1
- 125000006850 spacer group Chemical group 0.000 description 1
- 210000000952 spleen Anatomy 0.000 description 1
- 229960005322 streptomycin Drugs 0.000 description 1
- 230000004083 survival effect Effects 0.000 description 1
- 239000000725 suspension Substances 0.000 description 1
- 238000003786 synthesis reaction Methods 0.000 description 1
- 238000012385 systemic delivery Methods 0.000 description 1
- 238000011287 therapeutic dose Methods 0.000 description 1
- 201000002510 thyroid cancer Diseases 0.000 description 1
- 210000001685 thyroid gland Anatomy 0.000 description 1
- 208000013076 thyroid tumor Diseases 0.000 description 1
- 238000010361 transduction Methods 0.000 description 1
- 230000026683 transduction Effects 0.000 description 1
- 238000012250 transgenic expression Methods 0.000 description 1
- 230000001960 triggered effect Effects 0.000 description 1
- 239000001226 triphosphate Substances 0.000 description 1
- GPRLSGONYQIRFK-MNYXATJNSA-N triton Chemical compound [3H+] GPRLSGONYQIRFK-MNYXATJNSA-N 0.000 description 1
- 101150079396 trpC2 gene Proteins 0.000 description 1
- 229960005486 vaccine Drugs 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K35/00—Medicinal preparations containing materials or reaction products thereof with undetermined constitution
- A61K35/12—Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
- A61K35/48—Reproductive organs
- A61K35/51—Umbilical cord; Umbilical cord blood; Umbilical stem cells
-
- 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
- A61K2239/26—Universal/off- the- shelf cellular immunotherapy; Allogenic cells or means to avoid rejection
-
- 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
- A61K2239/46—Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
- A61K2239/58—Prostate
-
- 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
- A61K39/12—Viral antigens
-
- 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
- A61K39/39—Medicinal preparations containing antigens or antibodies characterised by the immunostimulating additives, e.g. chemical adjuvants
-
- 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
- A61K39/4611—T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
-
- 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
- A61K39/462—Cellular immunotherapy characterized by the effect or the function of the cells
- A61K39/4621—Cellular immunotherapy characterized by the effect or the function of the cells immunosuppressive or immunotolerising
-
- 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
- A61K39/46433—Antigens related to auto-immune diseases; Preparations to induce self-tolerance
-
- 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
- A61K39/46434—Antigens related to induction of tolerance to non-self
-
- 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
- A61K39/464493—Prostate associated antigens e.g. Prostate stem cell antigen [PSCA]; Prostate carcinoma tumor antigen [PCTA]; Prostatic acid phosphatase [PAP]; Prostate-specific G-protein-coupled receptor [PSGR]
-
- 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
- A61K39/464838—Viral antigens
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- 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
-
- 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
- C12N5/0636—T lymphocytes
- C12N5/0638—Cytotoxic T lymphocytes [CTL] or lymphokine activated killer cells [LAK]
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2319/00—Fusion polypeptide
- C07K2319/01—Fusion polypeptide containing a localisation/targetting motif
- C07K2319/03—Fusion polypeptide containing a localisation/targetting motif containing a transmembrane segment
-
- 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
- C12N2710/00—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
- C12N2710/00011—Details
- C12N2710/16011—Herpesviridae
- C12N2710/16111—Cytomegalovirus, e.g. human herpesvirus 5
- C12N2710/16134—Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
-
- 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
- C12N2710/00—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
- C12N2710/00011—Details
- C12N2710/16011—Herpesviridae
- C12N2710/16211—Lymphocryptovirus, e.g. human herpesvirus 4, Epstein-Barr Virus
- C12N2710/16234—Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
Definitions
- the present invention relates generally to methods of culturing cells, and more specifically to culturing cells for cell therapy. It further relates to the production of T cells with therapeutic attributes for use in Adoptive Cell Therapy.
- the desired cells are a relatively small population within a composition of cells that are placed into cell culture devices.
- the composition of cells typically includes the source of the desired cells (such as peripheral blood mononuclear cells), feeder cells that stimulate growth of the desired cells, and/or antigen presenting.
- Culture devices and methods that allow the medium that cells reside in to be in a generally undisturbed state are favored since the cells remain relatively undisturbed. Such devices include standard tissue culture plates, flasks, and bags.
- the culture progresses in stages generally consisting of allowing the cell composition to deplete the medium of growth substrates such as glucose, removing the spent medium, replacing the spent medium with fresh medium, and repeating the process until the desired quantity of desired cells is obtained.
- the cell composition is moved to other devices to initiate a new stage of production as the desired cell population increases and additional growth surface is needed.
- the rate of population growth of the desired cells slows as the population of cells upon the growth surface increases. The end result is that it is very time consuming and complicated to produce a sizable population of desired cells.
- EBV-CTLs Epstein Barr virus
- the conventional method for optimal expansion of EBV-CTLs uses standard 24-well tissue culture plates, each well having 2 cm of surface area for cells to reside upon and the medium volume restricted to 1 ml/cm 2 due to gas transfer requirements.
- the culture process begins by placing a cell composition comprised of PBMC (peripheral blood mononuclear cells) in the presence of an irradiated antigen presenting cell line, which may be a lymphoblastoid cell line (LCL), at a surface density (i.e.
- EBV-CTLs are selectively expanded again in the presence of irradiated antigen presenting LCL at a new surface density ratio of 4: 1, with a minimum surface density of about 2.5xl0 5 EBV-CTL/cm 2 .
- Medium volume is limited to a maximum ratio of 1 ml/cm 2 of growth surface area to allow oxygen to reach the cells, which limits growth solutes such as glucose.
- the maximum surface density that can be achieved is about 2x10 6 EBV-CTL/cm 2 .
- the maximum weekly cell expansion is about 8-fold (i.e. 2x10 6 EBV-CTL/cm 2 divided by 2.5x10 5 EBV-CTL/cm 2 ) or less.
- Continued expansion of EBV-CTLs requires weekly transfer of the EBV-CTLs to additional 24-well plates with antigenic re-stimulation, and twice weekly exchanges of medium and growth factors within each well of the 24-well plate.
- EBV- CTLs The culture of EBV- CTLs is but one example of the complex cell production processes inherent to cell therapy. A more practical way of culturing cells for cell therapy that can reduce production time and simultaneously reduce production cost and complexity is needed.
- T cells with native antigen specificity i.e. T cells that are directed against a particular peptide derived from a specific target antigen when presented in the context of particular human leukocyte antigen (HLA) allele
- HLA human leukocyte antigen
- the first adoptive T cell transfer protocols in the allogeneic hematopoietic stem cell transplant (HSCT) setting were based on the premise that donor peripheral blood contained T cells able to mediate antitumor and/or antiviral activity in the HSCT recipient. Accordingly, donor lymphocyte infusions (DLI) have been extensively used to provide anti-tumor immunity, and to a lesser extent, antiviral immunity.
- HSCT allogeneic hematopoietic stem cell transplant
- DLIs should contain memory T cells specific for tumors as well as a broad range of viruses, however, while successful for the treatment of a proportion of infections with adenovirus and EBV, the efficacy of this therapy is limited by the low frequency of T cells specific for many common acute viruses (such as rotavirus (RSV) and parainfluenza) and the relatively high frequency of alloreactive T cells.
- RSV rotavirus
- the high ratio of alloreactive T cells to virus-specific T cells is especially problematic in recipients of haploidentical transplants, in whom a higher incidence of graft versus host disease (GVHD) limits the tolerable DLI dose, severely limiting the dose of virus-specific T cells received.
- GVHD graft versus host disease
- An alternative strategy to prevent and treat specific viral infections after HSCT is the adoptive transfer of ex vz ' vo-expanded T cells with antiviral activity.
- the specific expansion of virus-reactive T cells has the advantage of increasing the numbers of virus-specific T cells that can be infused without increasing alloreactive T cells.
- Infusion of enriched antigen-specific T cells with reactivity against a particular antigen potentially increases therapeutic potency while decreasing undesired off-target effects such as GVHD and this therapeutic modality has proven safe and effective for the treatment of hematological malignancies as well as solid tumors such as melanoma and EBV-associated malignancies such as Hodgkin's lymphoma and nasopharyngeal carcinoma.
- the therapeutic benefit itself depends on the use/administration of HLA- matched or partially matched T cells.
- MHC major histocompatibility complex
- TCR native T cell receptor
- the therapeutic benefit is mediated by the specific interaction of the native or natural T cell receptor with the target antigen.
- this interaction can only take place in a compatible HLA setting (i.e.
- the therapeutic attribute of the T cell that provides its therapeutic purpose is the native antigen specificity of the donor T cells.
- This inherent requires at least a partial HLA match between the donor and the recipient, and in the allogeneic setting creates the potential for off-target effects such as GVHD.
- Others are proposing elimination of the donor T cells antigen receptors altogether through complex genetic engineering, and re-engineering the T cells to carry chimeric antigen receptors, thereby eliminating all innate recognition capacity of the T cell. However, this further complicates the method of producing T cells, which is already one of the main problems of Adoptive Cell Therapy.
- the unconventional conditions include reduced surface density (i.e. cells/cm 2 ) of desired cells, novel ratios of desired cells to antigen presenting and/or feeder cells, and/or use of growth surfaces comprised of gas permeable material with increased medium volume to surface area ratios.
- Embodiments of this invention relate to improved methods of culturing cells for cell therapy applications. They include methods that reduce the time, cost, and complexity needed to generate a desired number of desired cells by use of various novel methods that allow the desired cell population to maintain a higher growth rate throughout the production process relative to conventional methods.
- One aspect of the present invention relies on conducting the culture process in stages and establishing conditions at the onset of one or more stages that allow the growth rate of the desired cell population to exceed what is currently possible. At least one stage of culture, and preferably nearly all, establish initial conditions that include the desired cells resting either on non-gas permeable or gas permeable growth surfaces at unconventionally low surface density and at an unconventional ratio of antigen presenting cells (and/or feeder cells) per desired cell.
- the desired cell population can experience more doublings in a shorter period of time than allowed by conventional methods, thereby reducing the duration of production.
- Another aspect of the present invention relies on conducting the culture process in stages and establishing conditions at the onset of one or more stages such that the growth rate of the desired cell population exceeds what is currently possible. At least one stage of culture, and preferably nearly all, establish conditions that include the desired cells resting on a growth surface comprised of gas permeable material at unconventionally high medium volume to growth surface area ratios.
- the desired cell population can experience more doublings in a shorter period of time than is allowed by conventional methods, thereby reducing the duration of production.
- Another aspect of the present invention relies on conducting the culture process in stages and establishing conditions of each stage such that the growth rate of the desired cell population exceeds what is currently possible. At least one stage of culture, and preferably nearly all, establish initial conditions that include the desired cells resting on growth surfaces comprised of gas permeable material at unconventionally low surface density (i.e. cells/cm 2 ) with an unconventional ratio of antigen presenting cells (and/or feeder cells) per desired cell and in the presence of unconventionally high medium volume to growth surface area ratios.
- the desired cell population can experience more doublings in a shorter period of time than conventional methods allow, thereby reducing the duration of production.
- allogeneic T-Vehicles are created with therapeutic attributes that have a therapeutic purpose that will benefit recipients while not exposing the recipient to graft-versus-host-disease (GVHD).
- GVHD graft-versus-host-disease
- a therapeutic treatment is undertaken by obtaining T- Vehicles that are created by a process comprising stimulating donor PBMCs or donor cord blood with an antigen in order to activate the growth of T cells that have native antigen specificity to the antigen(s). Doing so produces an antigen-specific T cell population that is comprised of native antigen receptors that have antigen specificity to the antigen(s) that were used to stimulate their growth.
- the antigen-specific T cell population is altered to include at least one therapeutic attribute which does not include the native antigen receptors and has a therapeutic purpose that is independent of the antigen specificity of the native antigen receptors, thereby creating a population of T- Vehicles.
- the T-Vehicles are then delivered to a recipient that can derive therapeutic benefit from the T-Vehicles, independent of whether or not the cells of the recipient present antigen recognized by the native antigen receptor(s) of the T-Vehicles and/or wherein the cells of the recipient do not present antigen recognized by the native antigen receptor(s) of the T-Vehicles
- a therapeutic treatment is undertaken by obtaining T-Vehicles that are created by a process comprising stimulating donor PBMCs or donor cord blood with an antigen in order to activate the growth of T cells that have native antigen specificity to the antigen(s). Doing so produces an antigen-specific T cell population that is comprised of native antigen receptors that have antigen specificity to the antigen(s) that were used to stimulate their growth.
- the antigen-specific T cell population is altered to include at least one therapeutic attribute which does not include the native antigen receptors and has a therapeutic purpose that is independent of the antigen specificity of the native antigen receptors, thereby creating a population of T-Vehicles.
- the T- Vehicles are then delivered to a recipient that can derive therapeutic benefit from the T-Vehicles and does not have an HLA match to the T- Vehicles.
- T-Vehicles are altered to become loaded with recombinant proteins administered as an adjuvant with immunotherapies, altered with the therapeutic attribute of chemotherapeutic agents for the targeted treatment of cancer, altered with the therapeutic attribute of antimicrobial agents, altered with the therapeutic attribute of expressing transgenic molecules that confer the cells with tumor specificity, altered with the therapeutic attribute of being loaded or engineered with recombinant proteins for the treatment of autoimmune diseases, altered to express suicide genes, and/or are altered with the therapeutic attribute of loaded and/or engineered to in- vivo imaging.
- a method of producing antigen specific T cells with desired antigen recognition is attained by placing PBMCs or cord blood into a cell culture device, adding more than one antigen into the cell culture device in order to activate the growth of more than one population of antigen specific T cells, each population capable of recognizing one of the antigens, allowing a period of time for the antigen specific T cells to initiate population expansion, assessing the culture to determine the presence and/or quantity of at least one population of antigen specific T cells, determining which of the populations of T cells is suitable for continued proliferation, and re-stimulating the culture only with antigens recognized by the suitable populations of T cells.
- T cells with desired antigen recognition is attained by placing PBMCs or cord blood into a cell culture device, initially adding more than one antigen into the cell culture device in order to activate the growth of more than one population of antigen specific T cells, each population capable of recognizing one of the antigens, allowing a period of time for the antigen specific T cells to initiate population expansion, separating the culture into more than one device, adding only one of the initial antigens into each device, and determining which of the devices contains a population of antigen specific T cells suitable for continued proliferation, and terminating the culture in devices that do not contain a population of antigen specific T cells suitable for continued proliferation.
- donor T cells are produced with native antigen specificity that only allows them to recognize a single eptitope of antigens that are not present on normal human cells and not present on normal mammalian cells.
- Figure 1A shows the population of antigen-specific T cells in Example 1 undergoes at least 7 cell doublings after the initial stimulation over the first 7 days.
- Figure IB shows data demonstrating the magnitude of expansion of a T cell population within a cell composition over time as determined by tetramer analysis for Example 1.
- Figure 1C the rate of population growth of antigen-specific T cells diminishes over a 23 day period in Example 1.
- Figure 2 shows a table that illustrates the discrepancy between the potential expansion and observed fold expansion of antigen-specific T cells in Example 1.
- Figure 3A shows the presence of antigen-specific T cells following stimulations in Example 2.
- Figure 3B shows the expansion of a population of antigen-specific T cells as surface densities diminish from lxl0 6 /cm 2 to 3.1xl0 4 /cm 2 while maintaining an antigen-specific T cell to antigen presenting cell ratio of 4: 1 in Example 2.
- Figure 3C shows the expansion of a population of antigen-specific T cells as surface densities diminish from lxl0 6 /cm 2 to 3.1xl0 4 /cm 2 while in the presence of a fixed number of antigen presenting cells in Example 2.
- Figure 4 shows an example of results obtained when continuing the work described in Figure 3, which further demonstrated that when desired cells need the support of other cells, unconventionally low desired cell surface density can initiate population expansion so long as desired cells are in the presence of an adequate supply of feeder and/or antigen presenting cells.
- Figure 5 shows a histogram demonstrating the ability to repeat the magnitude of the population expansion of desired cells by initiating culture at three differing cell surface densities (CTL/cm 2 ).
- Figure 6 shows a cross-sectional view of a gas permeable test fixture used to generate data.
- Figure 7A shows the growth curves of antigen-specific T cells produced in accordance with the present invention in comparison to conventional methods as undertaken in Example 5.
- Figure 7B shows that for Example 5, cell viability was significantly higher in antigen- specific T cells produced in accordance with the present invention in comparison to conventional methods as determined by flow cytometric forward vs. side scatter analysis.
- Figure 7C shows that for Example 5, cell viability was significantly higher in antigen- specific t cells produced in accordance with the present invention in comparison to conventional methods as determined by Annexin-PI 7AAD.
- Figure 7D showed that for Example 5, the superior growth of cells produced in the novel methods of the present invention exhibited the same cell specific growth rate as cell cultured using conventional methods as determined by daily flow cytometric analysis of CFSE labeled cells, confirming that the increased rate of cell expansion resulted from decreased cell death.
- Figure 8A shows how EVB-CTLs were able to expand beyond what was possible in conventional methods without need to exchange medium.
- Figure 8B shows how the culture condition of Example 6 did not modify the final cell product as evaluated by Q-PCR for EBER.
- Figure 8C shows how the culture condition of Example 6 did not modify the final cell product as evaluated by Q-PCR for B cell marker CD20.
- Figure 9 shows an illustrative example in which we experimentally demonstrated that a very low cumulative surface density of desired cells and antigen presenting cells (in this case AL-CTLs and LCLs cells combining to create a cell composition with a surface density of 30,000 cells/cm 2 ) was unable to initiate outgrowth of the AL-CTL population.
- Figure 10A presents data of Example 8 that show how two novel methods of culturing cells produce more cells over a 23 day period than a conventional method.
- Figure 10B shows a photograph of cells cultured in a test fixture in Example 8.
- Figure IOC shows that in Example 8, the two novel methods of culture and the conventional method all produce cells with the same phenotype.
- FIG. 10D shows that for Example 8, a representative culture in which T cells stimulated with EBV peptide epitopes from LMPl , LMP2, BZLFl and EBNAl of EBV and stained with HLA-A2-LMP2 peptide pentamers staining showed similar frequencies of peptide- specific T cells.
- Figure 10E shows that for the novel methods and the conventional method of Example 8, cells maintained their cytolytic activity and specificity and killed autologous EBV-LCL, with low killing of the HLA mismatched EBV-LCL as evaluated by 51 Cr release assays.
- Figure 11 shows a graphical representation of expansion of a desired cell population on a growth surface under the conventional scenario as compared to population expansion of the desired cell type using one aspect of the present invention.
- Figure 12 shows an example of the advantages that can be obtained by utilizing a growth surface comprised of gas permeable material and an unconventionally high medium volume to growth surface area ratio beyond 1 or 2 ml/cm 2 .
- Figure 13 shows a graphical representation of a novel method of expansion of a desired cell population on a growth surface under the conventional scenario as compared to population expansion of the desired cell type under one embodiment of the present invention in which the cell surface density at the completion of is much greater than conventional surface density.
- Figure 14 shows another novel method of cell production that provides yet further advantages over conventional methods.
- Figure 15 shows a comparison of each production method depicted in Figure 14 to demonstrate the power of the novel method and why it is useful to adjust the production protocol at various stages to fully capture the efficiency.
- Figure 16 shows an example of how one could adjust the production protocol in the novel method to gain efficiency as production progresses.
- Figure 17 shows test results demonstrating T- Vehicles are unable to recognize cells from mismatched allogeneic donors.
- Figure 18 shows test results indicating donor T cells can be altered to create T-Vehicles with the therapeutic attribute of CD34A-IL7 cytokine expression, as determined by flow analysis.
- Figure 19A shows test results indicating systemic delivery of IL7 cytokine results in more cytokine being detected on the kidney of mice than at their tumor site.
- Figure 19B shows test results indicating T-Vehicle delivery of IL7 cytokine results in greater cytokine concentration at the mice tumor sites, when compared with other organs, and shows how cytokine production was sustained at the tumor for at least 2 weeks after the administration of the T-vehicles.
- Figure 20 shows test results indicating donor T cells can be altered to create T-Vehicles with the therapeutic attribute of CAR-PSCA, as determined by flow analysis.
- Figure 21 shows test results indicating T- Vehicles with the therapeutic attribute of CAR-
- PSCA are able to eradicate tumor cells.
- Figure 22A shows T- Vehicles with receptors capable of binding IL4 are in proximity of tumor cells expressing IL4 cytokine.
- Figure 22B shows how T-Vehicles can bind IL4 cytokines, and the quantity of IL4 cytokines protecting the tumor cells can be greatly reduced.
- Figure 23 shows test results demonstrating T-Vehicles, having a therapeutic attribute of expressing extra-cellular recombinant cytokine receptors IL4R/7, are able to deplete IL4 cytokine.
- Figure 24A shows how T-Vehicles loaded with chemotherapeutic agent will migrate towards the site of inflammation.
- Figure 24B shows how the Recipient immune system will target the T-Vehicles, which are located at the site of the Tumor cells.
- FIG. 24C shows how, under attack by the Recipient immune system, T-Vehicles will release their payload, in this case a chemotherapeutic agent, at the site of the Tumor cells.
- Antigen presenting cells Cells that act to trigger the desired cells to respond to a particular antigen.
- Desired cells The specific type of cell that that the production process aims to expand in quantity.
- the desired cells are non-adherent and examples includedie regulatory T cells (Treg), natural killer cells (N K), tumor infiltrating lymphocytes (TIL), primary T lymphocytes and a wide variety of antigen specific cells, and many others (all of which can also be genetically modified to improve their function, in-vivo persistence or safety).
- feeder cells and/or antigen presenting cells that can include PBMC, PHA blast, OKT3 T, B blast, LCLs and K562, (natural or genetically modified to express and antigen and/or epitope as well as co-stimulatory molecules such as 41BBL, OX40, CD80, CD86, HLA, and many others) which may or may not be pulsed with peptide or other relevant antigens.
- EBV Epstein Barr Virus
- EBV-CTL A T cell that specifically recognized EBV-infected cells or cells expressing or presenting EBV-derived peptides through its T cell surface receptor.
- EBV-LCL Epstein Barr virus transformed B lymphoblastoid cell line.
- Feeder cells Cells that act to cause the desired cells to expand in quantity. Antigen presenting cells can also act as feeder cells in some circumstances.
- Growth surface The area within a culture device upon which cells rest.
- PBMCs Peripheral Blood Mononuclear Cells derived from peripheral blood, which are a source of some of the desired cells and which can act as feeder cells.
- Responder A cell that will react to a stimulator cell.
- Static cell culture A method of culturing cells in medium that is not stirred or mixed except for occasions when the culture device is moved from location to location for routine handling and/or when cells are periodically fed with fresh medium and the like. In general, medium in static culture is typically in a quiescent state. This invention is directed to static cell culture methods. Stimulated: The effect that antigen presenting and/or feeder cells have on the desired cells. Stimulator (S): A cell that will influence a responder cell.
- Surface density The quantity of cells per unit area of the surface within the device upon which the cells rest.
- EXAMPLE 1 Demonstration of limitations of conventional methods.
- the data of this example demonstrate the limits of conventional culture methods for the production of EBV-CTL in standard 24 well tissue culture plates (i.e. 2 cm 2 surface area per well) using a medium volume of 2 ml per well (i.e. medium height at 1.0 cm and a medium volume to surface area ratio of lml/cm ).
- Stage 1 of culture, day 0 The expansion of an EBV-CTL population was initiated by culturing a cell composition of PBMCs from normal donors (about lxl 0 6 cells/ml) with antigen presenting gamma-irradiated (40 Gy) autologous EBV-LCLs at a 40: 1 ratio (PBMC:LCLs) and a medium volume to growth surface ratio of 1 ml/cm 2 thereby establishing a cell composition surface density of about lxlO 6 cells/cm 2 in RPMI 1640 supplemented with 45% Click medium (Irvine Scientific, Santa Ana, CA), with 2 mM GlutaMAX-I, and 10% FBS.
- PBMC:LCLs 40: 1 ratio
- Click medium Irvine Scientific, Santa Ana, CA
- Stage 2 of culture, day 9-16 On day 9, EBV-CTLs were harvested from the cell composition created in Stage 1 , resuspended in fresh medium at a surface density of 0.5x10 6 EBV-CTL/cm 2 and re-stimulated with irradiated autologous EBV-LCLs at a ratio 4: 1 CTL:LCL (surface density 0.5xl0 6 CTL/cm 2 : 1.25x10 5 LCL/cm 2 ).
- IL-2 human IL-2
- Stage 3 of culture, day 17-23 The conditions of Stage 2 were repeated with twice weekly addition of IL-2 and the culture was terminated on day 23. Although the culture was terminated, it could have been continued with additional culture stages that mimicked that of stages 2 and 3.
- Cell surface Cells were stained with Phycoerythrin (PE), fluorescein isothiocyanate (FITC), periodin chlorophyll protein (PerCP) and allophycocyanin (APC)-conjugated monoclonal antibodies (MAbs) to CD3, CD4, CD8, CD56, CD 16, CD62L, CD45RO, CD45RA, CD27, CD28, CD25, CD44 from Becton-Dickinson (Mountain View, CA, USA). PE-conjugated tetramers (Baylor College of Medicine) and APC-conjugated pentamers (Proimmune Ltd, Oxford, UK), were used to quantify EBV-CTL precursor frequencies. For cell surface and pentamer staining 10,000 and 100,000 live events, respectively, were acquired on a FACSCalibur flow cytometer and the data analyzed using Cell Quest software (Becton Dickinson).
- PE Phycoerythrin
- FITC fluorescein isothio
- CFSE labeling to measure cell division To assess the doubling rate of 2 x 10 7 PBMC or EBV-specific CTLs (EBV-CTLs) were washed twice and resuspended in 850 ⁇ 1 lx phosphate -buffered saline (PBS) containing 0.1% Fetal Bovine Serum (FBS) (Sigma- Aldrich).
- PBS phosphate -buffered saline
- FBS Fetal Bovine Serum
- AimexinV-7-AAD staining To determine the percentage of apoptotic and necrotic cells in our cultures we performed Annexin-7-AAD staining as per manufacturers' instructions (BD Pharmingen tm #559763, San Diego, CA). Briefly, EBV-CTL from the 24-well plates or the G- Rex were washed with cold PBS, resuspended in IX Binding Buffer at a concentration of lxl 0 6 cells/ml, stained with Annexin V-PE and 7-AAD for 15 minutes at RT (25°C) in the dark. Following the incubation the cells were analyzed immediately by flow cytometry.
- Chromium release assay We evaluated the cytotoxic activity of EBV-CTLs in standard 4-hour 51 Cr release assay, as previously described. As desired cells we used autologous and HLA class I and II mismatched EBV-transformed lymphoblastoid cell line (EBV-LCL) to measure MHC restricted and unrestricted killing, as well as the K562 cell line to measure natural killer activity. Chromium-labeled desired cells incubated in medium alone or in 1 % Triton X- 100 were used to determine spontaneous and maximum 51 Cr release, respectively. The mean percentage of specific lysis of triplicate wells was calculated as follows: [(test counts -spontaneous counts)/(maximum counts— spontaneous counts)] x 100.
- Enzyme-Linked Immunospot (ELIspot) assay was used to quantify the frequency and function of T cells that secreted IFNy in response antigen stimulation.
- CTLs were resuspended at lxl0 6 /ml in ELIspot medium [(RPMI 1640 (Hyclone, Logan, UT) supplemented with 5% Human Serum (Valley Biomedical, Inc., Winchester, Virginia) and 2-mM L-glutamine (GlutaMAX-I, Invitrogen, Carlsbad, CA)].
- ELIspot medium (RPMI 1640 (Hyclone, Logan, UT) supplemented with 5% Human Serum (Valley Biomedical, Inc., Winchester, Virginia) and 2-mM L-glutamine (GlutaMAX-I, Invitrogen, Carlsbad, CA)].
- the population of antigen-specific T cells undergoes at least 7 cell doublings after the initial stimulation over the first 7 days, as shown in Figure 1A.
- a weekly T cell expansion of 128-fold (as measured by the frequency of antigen- specific T cells times the total number of cells in the cell composition).
- the frequency of tetramer positive cells after the first, second, and third stimulations is shown in Figure IB.
- RAK and QAK was 0.02% and 0.01%, respectively.
- the frequency of tetramer- positive T cells in the cell composition had increased from 0.02% and 0.01% to 2.7% and 1.25%, respectively.
- Example 1 demonstrates that the amount of time it takes to produce the desired cells is typically delayed after roughly the first week of production since the rate of population expansion of the desired cells decreases in subsequent stages of culture.
- EXAMPLE 2 Reducing the amount of time needed to increase the desired cell population can be achieved by reducing the cell surface density of the desired cell population as the onset of any given stage or stages of culture. We hypothesized that the decreased rate of expansion of the desired cell population following the second T cell stimulation compared to the first stimulation was due to limiting cell culture conditions that resulted in activation induced cell death (AICD).
- AICD activation induced cell death
- the EBV antigen-specific T cell component of PBMCs represents, at most, 2% of the population and so the antigen-specific responder T cell seeding density is less than 2xl0 4 per cm 2 , with the remaining PBMC acting as non-proliferating feeder cells (seen as the CFSE positive cells in Figure 3A) that sustain optimal cell-to-cell contact allowing proliferation of the antigen-specific CTLs.
- the majority of T cells are antigen-specific, and although the total cell density of the composition is about the same, the proliferating cell density is 50 to 100 fold higher.
- the majority of cells proliferate and may therefore rapidly consume and exhaust their nutrients and 0 2 supply.
- EXAMPLE 3 A minimum surface density of a cell population that includes the desired cells and/or antigen presenting cells can allow outgrowth of a desired cell population that is seeded at very low surface density.
- Figure 4 shows an example of results we obtained when continuing the work described in Figure 3, which further demonstrated that when desired cells need the support of other cells, unconventionally low desired cell surface density can initiate population expansion so long as desired cells are in the presence of an adequate supply of feeder and/or antigen presenting cells.
- a total cell composition with a surface density and R:S ratio of between about l .OxlO 6 desired cells/cm 2 at an R:S ratio of 8 to 1 and merely about 3900 desired cells/cm 2 at an R:S ratio of 1 to 32 could allow desired cells to be greatly expanded to over 50 fold times the starting surface density, at which point we discontinued testing.
- EXAMPLE 4 The ability to allow a production process to repeat in stages by initiating a stage with an unconventionally low desired cell surface density, allowing population expansion, terminating the stage and repeating conditions was demonstrated to deliver repeatable outcomes.
- Example 3 We continued the assessments described in Example 3 at three of the desired cell surface densities (CTL/cm 2 ) as shown in Figure 5. Each specific seeding density was able to consistently attain the same fold expansion. The implications will be described in more detail further on as they relate to the ability to dramatically reduce the production time for a desired cell population.
- EXAMPLE 5 Culturing desired cells on a growth surface that is comprised of gas permeable material while simultaneously increasing the medium volume to growth surface area ratio increases the number of times a desired cell population can double in a given stage of culture relative to conventional methods and increases the surface density that is attainable.
- G-Rex Test fixtures
- EBV-specific CTL and irradiated autologous EBV-LCLs at the conventional 4: 1 ratio of CTL:LCL were cultured in G-Rex40 devices.
- EBV-CTLs in the G-Rex40 had increased from 5x10 /cm to a median of 7.9xl0 6 /cm 2 (range 5.7 to 8.1xl0 6 /cm 2 ) without any medium exchange.
- EBV-CTLs cultured for 3 days in conventional 24-well plates only increased from a surface density of 5xl0 5 /cm 2 to a median of 1.8xl0 6 /cm 2 (range 1.7 to 2.5xl0 6 /cm 2 ) by day 3.
- surface density could be further increased by replenishing medium whereas cell surface density could not be increased by replenishing medium or IL2 in the 24-well plate.
- EBV- CTL surface density further increased in the G-Rex40 to 9.5xl0 6 cells/cm 2 (range 8.5 xlO 6 to 11.0 xl0 6 /cm 2 ) after replenishing the medium and IL-2 on day 7 (data not shown).
- T cells were labeled with CFSE on day 0 and divided between a G-Rex40 device with a 40 ml medium volume and a 24 well plate with each well at a 2 ml medium volume.
- Daily flow cytometric analysis demonstrated no differences in the number of cell divisions from day 1 to day 3. From day 3 onwards, however, the population of desired cells cultured in the G-Rex40 continued to increase at a rate that exceeded the diminishing rate of the 2 ml wells, indicating that the culture conditions had become limiting as shown in Figure 7D.
- EXAMPLE 6 By use of unconventionally high ratios of medium volume to growth surface area and use of growth surfaces comprised of gas permeable material, the need to feed culture during production can be reduced while simultaneously obtaining unconventionally high desired cell surface density.
- G-Rex2000 refers to device as described in Figure 8, the exception being the bottom is comprised of a 100 cm 2 growth surface area and a 2000 ml medium volume capacity is available.
- EBV-LCLs were cultured in and expand in the G-Rex2000 without changing the cell phenotype.
- EBV-LCL were plated into a G-Rex2000 at a surface density of lxl 0 5 cells/cm 2 along with 1000 ml of complete RPMI medium to create a medium volume to surface area ratio of 10 ml/cm 2 .
- EBV-LCL were plated into a T175 flask at a surface density of 5x10 5 cells/cm 2 along with 30 ml of complete RPMI medium to create a medium volume to surface area ratio of about 0.18 ml/cm 2 .
- the EBV-LCL cultured in G-Rex2000 expanded more than those in the T175 flask without requiring any manipulation or media change. This culture condition did not modify the final cell product as evaluated by Q-PCR for EBER and B cell marker CD20 as presented in Figure 8B and Figure 8C.
- EXAMPLE 7 When sufficient feeder and/or antigen cells are not present at the onset of culture, desired cells may not expand. However, the cell composition can be altered to include an additional cell type acting as feeder cells and/or antigen presenting cell to allow expansion.
- Figure 9 shows an illustrative example in which we experimentally demonstrated that a very low cumulative surface density of desired cells and antigen presenting cells (in this case AL-CTLs and LCLs cells combining to create a cell composition with a surface density of 30,000 cells/cm 2 ) was unable to initiate outgrowth of the AL-CTL population.
- AL-CTLs and LCLs cells combining to create a cell composition with a surface density of 30,000 cells/cm 2
- this same cell composition could be made to grow by altering the composition to include another cell type acting as a feeder cell.
- the additive surface density of the antigen presenting cells and/or feeder cells and the desired cells should preferably be at least about 0.125xl0 6 cells/cm 2 to create enough surface density in the cell composition to initiate the expansion of the desired cell population.
- the use of growth surfaces comprised of gas permeable material was used in this example along with a medium volume to surface area ratio of 4 ml cm .
- EXAMPLE 8 Reduced desired cell surface densities, altered responder cell to stimulatory cell ratios, increased medium to growth surface area ratios, and periodic distribution of cells at a low surface density culture onto growth surfaces comprised of gas permeable material allow more desired cells to be produced in a shorter period of time and simplifies the production process when compared to other methods.
- G-Rex500 refers to device as described in Figure 6, the exception being the bottom is comprised of a 100 cm 2 growth surface area and a 500 ml medium volume capacity is available.
- a second stage was initiated on day 9, wherein lxlO 7 responder T cells were transferred from the G-Rex40 to a G-Rex500 test fixture.
- stage two of culture 200 ml of CTL medium was placed in the G-Rex500, creating a medium volume to surface area ratio at the onset of stage two of 2 ml/cm medium height at 2.0 cm above the growth surface area.
- the surface density of desired cells at the onset of stage two was lxlO 5 CTL/cm 2 with antigen presenting cells at a surface density of 5xl0 5 LCL/cm 2 , thereby creating a non-conventional 1 :5 ratio of desired cells to antigen presenting cells.
- This stage two cell surface density and R:S ratio produced consistent EBV-CTL expansion in all donors screened.
- IL-2 50U/ml - final concentration
- 200 ml of fresh medium bringing medium volume to surface area ratio to 4 ml/cm 2 .
- the cells were harvested and counted.
- the median surface density of CTLs obtained was 6.5xl0 6 per cm 2 (range 2.4xl0 6 to 3.5xl0 7 ).
- the use of growth surfaces comprised of gas permeable material allows increased medium volume to surface area ratios (i.e. greater than 1 ml/cm 2 ), lower cell surface densities (i.e.
- FIG. 10A shows the comparison of this G-Rex approach of Example 8 to the use of conventional methods of Example 1 and the G-Rex approach described in Example 5.
- the conventional method needed 23 days to deliver as many desired cells as could be delivered in either G-Rex method in about 10 days.
- the G-Rex approach of Example 8 was able to produce 23.7 more desired cells than the G-Rex method of Example 5 and 68.4 times more desired cells than the conventional method of Example 1.
- the desired cells continued to divide until day 27-30 without requiring additional antigen presenting cell stimulation provided the cultures were split when cell surface density was greater than 7xl0 6 /cm 2 .
- FIG. 10D shows a representative culture in which T cells stimulated with EBV peptide epitopes from LMP1, LMP2, BZLF1 and EBNA1 and stained with HLA-A2-LMP2 peptide pentamers staining showed similar frequencies of peptide-specific T cells. Further, the expanded cells maintained their cytolytic activity and specificity and killed autologous EBV- LCL (62% ⁇ 12 vs.
- Examples 1 - 8 have been presented to demonstrate to skilled artisans how the use of various conditions including reduced surface density of the desired cell population at the onset of a production cycle, reduced surface density ratios between responder cells and stimulating cells, growth surfaces comprised of gas permeable materials, and/or increased medium volume to growth surface area ratios can be used to expedite and simplify the production of cells for research and clinical application of cell therapy.
- Examples 1 - 8 were related to the production of antigen specific T cells, these novel culture conditions can be applied to many important suspension cell types with clinical relevance (or required for pre-clinical proof of concept murine models) including regulatory T cells (Treg), natural killer cells (N K), tumor infiltrating lymphocytes (TIL), primary T lymphocytes, a wide variety of antigen specific cells, and many others (all of which can also be genetically modified to improve their function, in-vivo persistence or safety).
- regulatory T cells Teg
- N K natural killer cells
- TIL tumor infiltrating lymphocytes
- primary T lymphocytes a wide variety of antigen specific cells, and many others (all of which can also be genetically modified to improve their function, in-vivo persistence or safety).
- Cells can be expanded with feeder cells and/or antigen presenting cells that can include PBMC, PHA blast, OKT3 T, B blast, LCLs and K562, (natural or genetically modified to express and antigen and/or epitope as well as co-stimulatory molecules such as 41BBL, OX40L, CD80, CD86, HLA, and many others) which may or may not be pulsed with peptide and/or a relevant antigen.
- Unconventionally Low Initial Surface Density One aspect of the present invention is the discovery that production time can be reduced relative to conventional methods by the use of lower desired cell surface density. In this manner, desired cells are able to have a greater numerical difference between their minimum and maximum cell surface densities than conventional methods allow. Preferably, when the rate of desired cell population growth has begun to diminish, but the quantity of desired cells is not yet sufficient to terminate production, the desired cells are re-distributed upon additional growth surfaces comprised of gas permeable material at low starting surface density once again.
- Figure 11 shows a graphical representation of expansion of a desired cell population on a growth surface under the conventional scenario as compared to population expansion of the desired cell type using one aspect of the present invention.
- the surface density of desired cells at the onset of a production stage is less than conventional surface density.
- this explanation does not describe the process of initially obtaining the desired cell population.
- the 'Day" of culture starts at "0" to allow skilled artisans to more easily determine the relative time advantages of this novel method.
- each production cycle of the conventional method begins at a conventional surface density of 0.5x10 6 desired cells/cm 2 while each production cycle of this example begins at a much lower and unconventional surface density of 0.125xl0 6 desired cells/cm 2 .
- 4 times more surface area i.e. 500,000/125,000
- the desired cells of the conventional method reaches a maximum surface density of 2xl0 6 cells/cm 2 in 14 days.
- 1 cm 2 of growth area delivers 2x10 6 cells/cm 2 which are then re-distributed onto 4 cm 2 of growth area so that production can be continued using the conventional starting density of 0.5x10 6 cells/cm 2 (i.e.
- the novel method depicted in Figure 11 instead of using the conventional method of depositing 500,000 desired cells onto 1 cm at the onset of production, distributes the 500,000 cells equally onto 4 cm of growth area to create at unconventionally low starting surface density of 125,000 desired cells/cm 2 on Day 0.
- the novel method as with the conventional method, has its growth rate about to diminish on Day 7.
- Cells in the novel method are at a surface density of lxlO 6 cells/cm 2 .
- this stage of culture has produced 4x10 6 cells that are then re-distributed onto 32 cm 2 of growth area so that production in Stage 2 can be continued using the starting surface density of 0.125xl 0 6 cells/cm 2 (i.e.
- the advantage of this aspect of the present invention is the production time reduction resulting from the reduction of cell surface density below that of conventional cell surface density in any particular application, wherein the particular conventional surface density used in this illustrative example may vary from application to application.
- Desired cells should be deposited upon a growth surface at an unconventionally low cell surface density such that:
- the desired cells are in the presence of antigen presenting cells and/or feeder cells and with medium volume to surface area ratio of up to 1 ml cm if the growth surface is not comprised of gas permeable and up to 2 ml/cm 2 if the growth surface is comprised of gas permeable, and b. the preferred surface density conditions at the onset of a production cycle being such that the target cell surface density is preferably less than 0.5x10 6 cells/cm 2 and more preferably diminishing as described in Figure 4, and
- the surface density of the desired cells plus the surface density of the antigen presenting cells and/or feeder cells is preferably at least about 1.25 xlO 5 cells/cm 2 .
- growth surfaces comprised of gas permeable material and higher medium volume to growth surface area ratios can simplify and shorten production.
- Another aspect of the present invention is the discovery that the use of growth surfaces comprised of gas permeable material and medium volume to growth surface area ratios that exceed conventional ratios, and repeated cycles of production that increase the amount of growth surface area used over time will reduce production duration.
- Figure 12 augments the discussion to show an example of the advantages that can be obtained by utilizing a growth surface comprised of gas permeable material and an unconventionally high medium volume to growth surface area ratio beyond 1 or
- FIG. 12 shows two production processes, labeled “conventional method” and “novel method.” At the onset of growth, each process begins with desired cells at a surface density of 0.5xl0 6 /cm 2 . However, the growth surface of in the novel method is comprised of gas permeable material and medium volume to
- the novel method can be terminated prior to time "X" with more cells produced than the conventional method, can be terminated at time “X” with about 1.5 times more cells produced than the conventional method, or can continue until the medium is depleted of nutrients with 2 times many desired cells produced as the conventional method in twice the time but without any need to handle the device for feeding.
- the conventional method In order for the conventional method to gather as many cells, the cells must be harvested and the process reinitiated, adding labor and possible contamination risk. Since cell therapy applications typically only are able to start with a fixed number of cells, the conventional method does not allow the option of simply increasing surface area at the onset of production.
- Figure 13 continues the example of Figure 12 to show how more than one production cycle can be of further benefit.
- Figure 13 shows a graphical representation of expansion of a desired cell population on a growth surface under the conventional method as compared to population expansion of the desired cell type under one novel method of the present invention in which the surface density of the novel method exceeds surface density of the conventional method.
- the 'Day" of culture starts at "0" to allow skilled artisans to more easily determine the relative time advantages of this aspect of the invention.
- both cultures are initiated using conventional desired cell surface density of 0.5x10 5 cells/cm 2 at "Day 0".
- the growth surface of the conventional method is also comprised of gas permeable material.
- the medium volume to growth surface ratio in the conventional method is 1 ml cm 2 as opposed to 4 ml/cm 2 in the novel method.
- the desired cell population in the conventional method begins to diminish in growth rate when it is at a surface density of about 1.5xl0 6 cells/cm 2 in about 4 days and reaches a maximum surface density of 2xl0 6 cells/cm 2 in 14 days.
- the desired cell population is distributed to 4 cm 2 of growth area at a surface density of 0.5xl0 6 /cm 2 in fresh medium at 1.0 ml/cm 2 and the production cycle begins again, reaching a surface density of 2xl0 6 cells/cm 2 in another 14 days and delivering 8xl0 6 desired cells in 28 days.
- the desired cell population in the novel method begins to diminish in growth rate when it is at a surface density of about 3x10 6 cells/cm 2 in roughly about 10 to 1 1 days and could reach a maximum surface density of 4xl0 6 cells/cm 2 in 28 days.
- the cycle ends when the desired cell population is still in a high rate of growth.
- the 3x10 6 cells are re-distributed to 6 cm 2 of growth surface area at a surface density of 0.5x10 6 /cm 2 in fresh medium at 4.0 ml/cm 2 and the production cycle begins again, with the desired cell population reaching a surface density of 3x10 6 cells/cm 2 in roughly another 10 to 11 days and delivering 18xl0 6 desired cells around 21 days.
- the novel method has produced over 2 times the number of desired cells as compared to the conventional method.
- Figure 14 shows another novel method in which still further advantages relative to conventional methods are obtained.
- skilled artisans will recognize that the description herein does not limit the scope of this invention, but instead acts to describe how to attain advantages of improved production efficiency.
- desired cells are doubling weekly in conventional conditions.
- the 'Day" of culture starts at "0" to allow skilled artisans to more easily determine the relative time advantages of this embodiment.
- issues previously described related to feeder and/or antigen presenting cell surface density ratios are not repeated to simplify this example.
- the conventional method begins with a surface density of 0.5xl0 6 cells/cm 2 and a medium volume to surface area ratio of 1 ml/cm 2 .
- the novel method of this example begins with a surface density of 0.06x10 6 cells/cm 2 , a growth surface area comprised of gas permeable material, and a medium volume to surface area ratio of 6 ml/cm 2 .
- a surface density of 0.06x10 6 cells/cm 2 a growth surface area comprised of gas permeable material
- a medium volume to surface area ratio of 6 ml/cm 2 a medium volume to surface area ratio of 6 ml/cm 2 .
- the population is determined to be reaching plateau from noting that plateau is initiated in the conventional method when cell surface density approaches 1.5 times the medium volume to surface area ratio (i.e. about 1.5xl0 6 cells/ml).
- a surface density of about 4.5xl0 6 cells/cm 2 at about 9 days cells are distributed onto 36 cm 2 of growth surface area and the production cycle begins anew.
- Figure 15 tabulates a comparison of each production method depicted in Figure 14, and extends to stages to demonstrate the power of the novel method, and why it is wise to adjust the production protocol at various stages to fully capture the efficiency.
- the novel method overpowers the conventional method after completing just the second stage of the production cycle, delivering nearly 1.37 times more cells in only about half the time with just 61% of the surface area requirement.
- the third stage of the production cycle creates a massive increase in cells and a corresponding increase in surface area.
- Figure 16 shows an example of how one could alter variables in the novel method to gain efficiency as production progresses.
- an increase in the starting surface density of cycle 3 from 0.06 to 0.70 cell/cm and a change to the final surface density from 4.5 to 7.5 cells/cm 2 can be undertaken.
- Increasing the final surface density is a matter of increasing the medium volume to surface area ratio beyond the initial 6 ml/cm 2 to a greater number. The greater the medium volume to surface area, the longer the cycle remains in rapid growth phase (i.e. the population expansion prior to plateau). In this case we have allowed 5 extra days to complete the rapid growth phase and raised the medium volume to surface area ratio to about 8 ml/cm .
- the target cell surface density is less than the conventional density, preferably at between about 0.5xl0 6 desired cells/cm 2 and about 3900 desired cells/cm 2 and total number of desired cells and antigen presenting cells and/or feeder cells being at least about 1.25xl0 5 cells/cm 2 , and
- T-Vehicles are comprised of a population of T cells that do not carry inherent risk of GVHD, further altered to include one or more therapeutic attributes capable of acting with a therapeutic purpose in order to provide recipients with a therapeutic benefit. Since T-Vehicles do not have a native capacity to initiate GVHD disease, they become an ideal biological transportation vehicle to arm with any number of weapons capable of fighting a wide variety of medical conditions and diseases.
- the present invention discloses methods for producing and using T-Vehicles that are armed with therapeutic attributes for the purpose of providing recipients the health benefits of Adoptive Cell Therapy without inherent risk of GVHD that is present in state-of-the-art methods.
- T- Vehicles function contrary to state-of-the-art methods for Adoptive Cell Therapy, as the therapeutic purpose of T-Vehicles is wholly unrelated to the native T cell receptor's antigen specificity.
- Skilled artisans are encouraged to recognize throughout the disclosures and illustrative embodiments presented, the therapeutic attribute of T-Vehicles does not include the native antigen receptors of the T-Vehicles.
- T-Vehicles are produced by stimulating donor PBMCs or donor cord blood with antigen in order to activate growth of donor T cells that have native antigen specificity to the antigen, thereby producing an antigen-specific T cell population that comprises antigen receptors with antigen specificity to the antigen.
- antigens that are not present on normal cells By selecting antigens that are not present on normal cells, a population of T cells with antigen receptors that are not able to recognize normal cells can be created.
- a population of T-Vehicles can be created that have a purpose independent of their antigen specific recognition and are not inherently prone to, or even capable of, initiating GVHD.
- T- Vehicles may encompass more than one population of native antigen-specific T cells, since T- Vehicles do not rely on their native antigen specificity for its therapeutic purpose, T- Vehicles can be infused into a recipient independent of whether or not the serotype of the recipient exhibits a positive match to any of the native antigen receptor(s) of T-Vehicles. Also, key attributes of T-Vehicles include their ability to be used in a HLA mismatched setting or, since the native T cell population(s) from which T-Vehicles are derived do not carry inherent risk of GVHD.
- T-Vehicles This allows allogeneic banks of T-Vehicles to be established that can service a wide segment of society without the limitations of HLA matching that is required in state-of-the- art methods.
- native T cells receptors of the T-Vehicles are incapable of recognizing cells in the recipient and initiating GVHD.
- T-Vehicles commence with their therapeutic activity in a completely HLA mismatched setting because they have been altered with therapeutics attributes that do not rely on the native antigen receptors to accomplish its therapeutic purpose.
- T-Vehicles are not limited to use in HLA mismatched setting however.
- T-Vehicles comprised of T cells that have native antigen receptors with highly restricted antigen-specificity against antigens not expressed on normal cells
- the initiation of GVHD disease can be avoided despite a partial HLA match between the recipient and the native antigen specificity of the T- Vehicle.
- the native antigen specificity of the T-Vehicles only allows them to recognize antigens that are not present on normal cells, more preferably normal human cells, even more preferably are only able to recognize a single epitope of antigens that are not present on normal mammalian cells.
- each dose of T-Vehicles differs in HLA so that the patient's immune system needs to re -prime itself each time it prepares to attack a new dose of T-Vehicles, thereby keeping the interval between each dose of T-Vehicles roughly equal.
- T cells that have native antigen receptors with highly restricted antigen-specificity Historically, producing populations of T cells at the scale needed for wide spread use in Adoptive Cell Therapy has been virtually impossible. State-of-the-art production methods for expanding T cells populations into suitably sized therapeutic doses are so impractical and unmanageable that they limit cell therapy to just very small population that must be treated at a small number of highly specialized institutes. A fundamental attribute of T- Vehicles is that their native T cell characteristics do not inherently expose the recipient to GVHD.
- the native antigen specificity of the T-Vehicles only allows them to recognize antigens that are not present on normal cells, more preferably normal human cells, even more preferably are only able to recognize a single epitope of antigens that are not present on normal mammalian cells, efficient production of these cells becomes a cornerstone for wide spread use of methods involving T-Vehicles.
- T cells are only present at very low, and sometimes undetectable, frequencies in donor PBMCs or cord blood.
- the problems inherent to state-of-the-art T cell production methods are compounded when trying to generate populations of T cells that are most suitable for use in T-Vehicles.
- PBMCs or cord blood i.e. the original pool of antigen specific T cells
- T cell population each population expressing an antigen receptor to one of the antigens presented.
- the intent is to subsequently select the most prolific and/or desirable native T cell population for production and terminate the others.
- the various T cell populations responding to the various antigens are likely to exhibit differing levels of population expansion, depending on the magnitude of their original population. Furthermore, some or all may continue to be undetectable.
- the culture is assessed for acceptable outgrowth of T cell populations reacting to any of the selected antigens.
- Such an assessment could be for just one population specific to one antigen, or to additional populations specific to additional antigens. If one antigen-specific T cell population is demonstrating acceptable expansion, re-stimulating that particular T cell population by only adding the antigen it recognizes into the device will cause the remaining T cells to eventually die, while the particular desired T cell population continues to proliferate.
- the culture can be re-stimulated with only the antigens those particular T cell populations are reacting to (thereby terminating expansion of less prolific T cell populations) or 2) the culture can split into more than one culture device, each device receiving a single antigen differing from all other devices antigen thereby causing only one T cell population to proliferate in each device with all but the most prolific cultures eventually being terminated.
- all culture devices are gas permeable and of the types described in co-pending U.S. Publication Nos. 2005/0106717 Al to Wilson et al. (hereinafter referred to as Wilson '717) and 2008/0227176 Al to Wilson (hereinafter referred to as Wilson ⁇ 76), which are both incorporated by reference herein, and rely on the methods of Vera '700.
- a population of PBMCs residing in a culture device could be presented with antigen A, antigen B, and antigen C. After period of time, the culture could be assessed for the presence and/or proliferation of populations reactive to antigens A, B, or C. If an antigen specific population reactive to antigen A is the only population not exhibiting acceptable frequencies and/or population expansion, it can be terminated by re-stimulation with only antigen B and antigen C. Alternatively, if antigen specific population reactive to antigen B and antigen C were proliferating about equally, but it was uncertain which would continue to proliferate the at best rate, the culture could be split into two devices with the expectation that one device would eventually continue production while the other would be terminated.
- the first device would receive antigen B and the second device would receive antigen C.
- T cells exhibiting antigen specificity to antigen B would proliferate in the first device but T cells exhibiting antigen specificity to antigen C eventually would die off.
- Examination of the frequency and/or population size could be undertaken with the intent of terminating the culture with the least efficient expansion of the desired T cell population.
- Skilled artisans are encouraged to recognize that a primary advantage of initiating culture with multiple antigens at onset, as opposed to just one antigen, is that it increases the prospects of finding a T cell population of suitable antigen specificity and growth rate.
- using multiple antigens in one device instead of multiple devices with one antigen makes more efficient use of PBMCs or cord blood, medium, cytokines, laboratory space, labor, and bio-hazardous disposal space.
- the native antigen specificity of the T- Vehicles only allows them to recognize antigens that are not present on normal cells, more preferably normal human cells, even more preferably are only able to recognize a single epitope of antigens that are not present on normal mammalian cells, this is non-limiting and there are many suitable attributes of the native antigen receptors skilled artisans are encouraged to consider. Many options and characteristics are suitable.
- the native antigen specificity of T- Vehicles can be composed of more than one population of T cells with native antigen specificity.
- the native antigen specificity of the T-Vehicles can be against a whole antigen or a single epitope of self or a non-self antigens; reptiles, amphibians, fish, or birds; invertebrates such as sponges, coelenterates, worms, arthropods, mollusks, or echinoderms; bacteria, fungus, parasites, and sponges; viruses including but not limited to adenovirus, Epstein-Barr virus (EBV), Cytomegalovirus (CMV), Adenovirus (Adv), Respiratory Syncytial virus (RSV), human herpesvirus 6 (HHV6), human herpesvirus 7 (HHV7), BK virus, JC virus, Influenza, H1N1, parainfluenza, herpes simplex virus (HSV), Varicella Zoster Virus (VZV), Parvovirus B19, Coronavirus, Metanpneumo virus, Bocavirus, or KI virus/WU virus;
- Leen '870 antigen presenting cells
- APCs antigen presenting cells
- Monocytes Monocytes
- Macrophages B cells
- T cells T cells
- PBMCs artificial antigen presenting cells
- engineered k562 any of which are able to present the desired antigens to produce the desired antigen specificity of the native donor T cell population and thus the native antigen specificity of the T-Vehicles
- use of antigen for the induction of the desired immune response in the donor cells by use of cell lysate containing the desired antigen, purified protein containing the desired antigen, recombinant protein containing the desired antigen, plasmid DNA encoding for the desired antigen, plasmid RNA encoding the desired antigen describe, and/or peptid
- Production of the T cell population is preferably undertaken using the methods of Vera '700, and/or those presented herein, and most preferable they are undertaken utilizing gas permeable culture devices of the types described in Wilson '717 and/or Wilson ' 176. Skilled artisans are encouraged to recognize that various methods in the described body of work may be more or less appropriate depending on the specific objectives of each application.
- various surface densities, medium heights, medium volume to growth surface areas and the like can be utilized, as well as stimulation with cytokines such as IL2, IL15, IL21 , IL12, IL7, IL27, IL6, IL18 and/or IL4 and various frequencies and concentration, and use of repetitive in vitro stimulation using any source of antigen in combination with any of the methods of presenting the antigen is possible and can be undertaken with or without cell sorting by methods including by not limited to gamma capture, magnetic isolation, single cell cloning, and/or flow cytometry.
- EXAMPLE 9 T- Vehicles with native T cell receptors recognizing the CMV epitope NLV are unable to recognize non-autologous cell targets.
- Antigen specific T cells with native antigen specificity to NLV-CMV were expanded from a frequency of 0.03% in PBMCs to 87% in 12 days using the methods previously described. These cells were then placed in culture with cells from three HLA mismatched donors presenting the target CMV antigen's NLV peptide.
- Figure 17 shows how the T- Vehicles were unable to recognize cells from mismatched allogeneic donors whether or not they expressed the NLV peptide ("allol” and “allol pep”, “allo2” and “allo2 pep”, “allo3” and “allo3 pep”) despite their full functionality as demonstrated by the capacity to recognize and kill autologous cells presenting the NLV peptide ("Auto4 pep") and avoid killing autologous cells not presenting the NLV peptide ("Auto4").
- Selecting and creating the desired therapeutic attribute(s) There are a wide variety of options for altering the antigen specific T cell population to include at least one therapeutic attribute. Examples follow that are non-limiting, but intended to provide skilled artisans with recognition of how the choice of therapeutic attribute depends on the therapeutic purpose and why the therapeutic attribute and its therapeutic purpose are independent of the antigen specificity of the T-Vehicles native antigen receptors.
- EXAMPLE 10 T-vehicles loaded with recombinant proteins administered as an adjuvant with immunotherapies.
- Immunotherapies are a class of therapies which are designed to elicit or amplify an immune response in a patient. Examples including administration of vaccines designed to activate an immune response directed against tumor antigens expressed on cancer cells or delivery of ex vivo expanded T cells or NK cells. Recombinant proteins such as cytokines like IL2, IL7, GM- CSF, have been administered systemically in order to promote the growth, expansion, persistence and/or function of these cells in vivo but the systemic administration of some cytokines (e.g.
- IL2 has been associated with in vivo toxicity including severe mucositis, nausea, diarrhea, edema, respiratory distress, liver and renal dysfunctions, and the expansion of regulatory T cells that impair the function of the induced/infused T cells.
- Administration of T- vehicles loaded with recombinant proteins including cytokines can overcome such toxicities by migrating to the site of inflammation, and delivering these recombinant proteins directly at the site of inflammation (induced by the immunotherapy).
- T-vehicles can be used to target the delivery of such cytokines instead of the traditional unspecific systemic administration.
- experiments were undertaken to create T-Vehicles able to produce the cytokine IL7 and to express a truncated form of CD34A which can be used to detect the percentage of transduce cells as wells as selecting the transgenic population.
- donor T cells with 98% native antigen specificity for the NLV epitope of CMV virus were successful altered to create T-Vehicles with the therapeutic attribute of CD34A-IL7 cytokine expression as determine by flow analysis.
- Further testing demonstrated that only T-Vehicles modified with the retroviral vector (CD34A-IL7 cytokine) were capable of producing IL7, as detected by ELISA.
- mice were divided into two groups (5 animals per group). In Group 1 , tumor bearing mice were treated with 2000ng of IL7 cytokine administered systemically by IV. In Group 2, mice were treated with a single IV injection of 10E+06 T-Vehicles. Random subjects from each group were then sacrificed at week 1 and week 2 to evaluate by ELISA the IL7 cytokine concentration at different locations including the heart, liver, kidney, spleen, peritoneum, tumor and blood.
- Group 1 tumor bearing mice were treated with 2000ng of IL7 cytokine administered systemically by IV.
- mice were treated with a single IV injection of 10E+06 T-Vehicles. Random subjects from each group were then sacrificed at week 1 and week 2 to evaluate by ELISA the IL7 cytokine concentration at different locations including the heart, liver, kidney, spleen, peritoneum, tumor and blood.
- Figure 19A shows the IL7 cytokine accumulation in the various locations for Group 1.
- the IL7 cytokine ELISA analysis demonstrate that higher cytokine levels were detected on the kidney and they were below detection at the tumor site.
- Figure 19B shows the IL7 cytokine accumulation in the various locations for Group 2.
- the IL7 cytokine ELISA analysis demonstrate greater cytokine concentration at the tumor site when compare with other organs and cytokine production was sustained at the tumor for at least 2 weeks after the administration of the T-vehicles. Therefore, T-Vehicles were able to migrate to the tumor site and preferentially deliver the cytokine IL7 for a sustained period of time. This clearly demonstrates the ability of the T-vehicles, with a therapeutic attribute capable of delivering cytokine, provides superior therapeutic benefits when compared state-of-the-art methods of cytokine delivery that are administered systemically.
- T-Vehicles have a limited in-vivo presence, as indicated by the reduction in cytokine concentration from week 1 to week 2. This can be viewed as an additional benefit of T-Vehicles, as they do not remain in the recipient.
- additional doses of T-Vehicles would be administered as needed until therapeutic outcome is met, and without additional doses, the T- Vehicles would be purged from the recipient
- Donor T cells can be modified to create T-Vehicles with the therapeutic attribute being a chimeric antigen receptor (CAR) that targets a particular antigen.
- CAR chimeric antigen receptor
- the therapeutic purpose of the T-Vehicle is the destruction of prostate tumor cells.
- quadrant E2, 57.23% of the donor T cells with were successful altered to create T-Vehicles with the therapeutic attribute of CAR-PSCA as determined by flow analysis.
- T-vehicles expressing CAR-PSCA were able to nearly eradicate the entire population of PSCA positive tumor cells, while simultaneously demonstrating extraordinarily selection for the PSCA antigen by leaving the PSCA negative cells unharmed. This clearly demonstrates the T-Vehicles capacity to create a therapeutic benefit unrelated to its native antigen specificity.
- EXAMPLE 12 Donor T cells can be altered to create T- Vehicles with the therapeutic attribute being a receptor that is capable of depleting unwanted cytokines in the recipient.
- Tumor cells protect from the immune system by the production of immune-suppressive cytokines which repress the anti-tumor effect of the endogenous T cells.
- Donor T cells can be altered to create T-vehicles with the therapeutic attribute of expressing whatever particular cytokine receptors are needed to provide the therapeutic purpose of vacuuming the unwanted particular cytokines from the tumor, thereby having the therapeutic benefit of making the tumor environment more permissive to immunotherapy strategies.
- Figure 22A and Figure 22B show a representation of such a process. In the depiction of Figure 22A, T-Vehicles including the therapeutic attribute of receptors capable of binding IL4 are in proximity of tumor cells expressing IL4 cytokine.
- the T-Vehicles have bound IL4 cytokines and the quantity of IL4 cytokines protecting the tumor cells is greatly reduced.
- the therapeutic attribute, the therapeutic purpose, and the therapeutic benefit of the T- Vehicle does not include, and is independent of, the native antigen receptor of the T- Vehicle.
- Experiments were conducted to evaluate the capacity of T-Vehicles, having a therapeutic attribute of expressing extra-cellular recombinant cytokine receptors IL4R/7, to deplete IL4 cytokine.
- T-Vehicles were prepared by altering donor T cells with native specificity for the NLV epitope of the CMV virus.
- T-Vehicles were culture in a 24 well plate in a volume of 2 mis of media in the presence of 2000pg/ml of IL4 and compared the donor T cells. The concentration of the cytokine IL4 was then evaluated by ELISA at 24, 48 and 72hs. Results are shown in Figure 23. Clearly the T-Vehicles were able to meet their therapeutic purpose, as the reduction of the immune-suppressive tumor growth factor IL4 cytokine over a 72 hour period was striking. To the contrary, donor T cells (i.e. the histograms labeled "Unmodified T-vehicle") showed no capacity to reduce the presence of IL4.
- T- Vehicles can be equipped with in order to become capable of meeting a therapeutic purpose intended to provide a recipient with a therapeutic benefit.
- the disclosed possibilities are now augmented by several additional examples.
- T- Vehicles altered with the therapeutic attribute of chemotherapeutic agents for the targeted treatment of cancer A variety of different chemotherapeutic agents or anti-neoplastic drugs are used to treat different types of cancers including breast, prostate, pancreatic, liver, lung, brain, leukemia, lymphoma, melanoma, and myeloma. Most chemotherapy is delivered intravenously, although a number of agents can be administered orally, and subsequently circulates throughout the body. Chemotherapy agents act by killing cells that divide rapidly, one of the main properties of most cancer cells. This means that chemotherapy also harms cells that divide rapidly under normal circumstances (e.g. cells in the bone marrow, digestive tract, and hair follicles).
- T-Vehicles loaded with these drugs has the potential to offset these toxicities. This can occur by loading T-Vehicles with a chemotherapeutic agent, infusing them into a recipient, whereby they will migrate to sites of inflammation (cancer) down a chemotactic gradient. In this manner, the chemotherapeutic agent is placed in proximity of the tumor cells as opposed to being administered in a systemic manner to the recipient.
- the recipient immune system will mount an attack on the T-Vehicles, causing them to be destroyed, but not without releasing the chemo therapeutic agent at the site of the tumor cells.
- the payload i.e. chemotherapy drug
- the payload can be deposited directly at the target site rather than being administered in a systemic manner, thus reducing the off-target toxicities associated with chemotherapy.
- T-Vehicles loaded with chemotherapeutic agent migrate towards the site of inflammation (i.e. tumor cells) and due to the HLA mismatch between T-Vehicles and the Recipient cells, the native antigen receptors of the T-Vehicles does not recognize the Recipient cells, arriving at the Tumor cells without initiating GVHD.
- the Recipient immune system has targeted the T-Vehicles, which are located at the site of the Tumor cells.
- T-Vehicles under attack by the Recipient immune system, T-Vehicles have released the chemotherapeutic agent at the site of the Tumor cells, thereby avoiding the off target toxicities inherent to state-of-the-art methods of delivering chemotherapy.
- T-Vehicles altered with the therapeutic attribute of antimicrobial agents An antimicrobial is a substance that kills or inhibits the growth of microorganisms such as bacteria, fungi, or protozoans. These agents are typically administered systemically and can be delivered in a more targeted manner if loaded onto T-Vehicles which have the ability to home to sites of inflammation in order to deliver their payload.
- T-Vehicles altered with the therapeutic attribute of producing recombinant proteins administered as an adjuvant with immunotherapies:
- T-Vehicles can also be engineered using viral (e.g. adenovirus, retrovirus, lentivirus) or non-viral transfection approaches to transgenically express recombinant proteins including cytokines, chemokines, enzymes, tumor antigens and cytokine receptors which can also be designed to act as an adjuvant to other immunotherapeutic interventions in order to enhance T cell persistence, promote expansion, induce homing, etc.
- viral e.g. adenovirus, retrovirus, lentivirus
- non-viral transfection approaches to transgenically express recombinant proteins including cytokines, chemokines, enzymes, tumor antigens and cytokine receptors which can also be designed to act as an adjuvant to other immunotherapeutic interventions in order to enhance T cell persistence, promote expansion, induce homing, etc.
- T- Vehicles altered with the therapeutic attribute of expressing transgenic molecules that confer the cells with tumor specificity In the same way T-Vehicles can be modified with recombinant protein such as cytokines, T-Vehicles can also be engineered using viral (e.g. adenovirus, retrovirus, lentivirus) or non-viral transfection approaches to transgenically express chimeric T cell receptors (CARs).
- viral e.g. adenovirus, retrovirus, lentivirus
- CARs chimeric T cell receptors
- T-Vehicles altered with the therapeutic attribute of being loaded or engineered with recombinant proteins for the treatment of autoimmune diseases Autoimmune diseases arise from an inappropriate immune response of the body against substances and tissues normally present in the body. In other words, the immune system mistakes some part of the body as a pathogen and attacks its own cells. This may be restricted to certain organs.
- the administration of T-Vehicles loaded with recombinant proteins such as IL10, TGFB, IL13 cytokines which will suppress the inflammation can overcome such autoimmune effect by delivering these recombinant proteins directly at the site of inflammation, thus delivering the payload directly where required rather than dispensing the recombinant protein indiscriminately.
- T-Vehicles can be engineered to express suicide genes: To allow the rapid and complete elimination of infused cells, T-Vehicles can be incorporated with a safety switches or suicide genes, which can be triggered should toxicity occur.
- the best validated of the suicide genes is thymidine kinase from herpes simplex virus I (HSV-tk). This enzyme phosphorylates the nontoxic prodrug ganciclovir, which then becomes phosphorylated by endogenous kinases to GCV-triphosphate, causing chain termination and single-strand breaks upon incorporation into DNA, thereby killing dividing cells.
- HSV-tk herpes simplex virus I
- inducible Fas Fas-associated death domain-containing protein (FADD)
- FADD Fas-associated death domain-containing protein
- Caspase9 a synthetic drug that has proven safe in healthy volunteers.
- FKBP FK-binding protein
- CID chemical inducer of dimerization
- API 903 a synthetic drug that has proven safe in healthy volunteers.
- Administration of this small molecule results in cross-linking and activation of the proapoptotic target molecules. Up to 90% of T cells transduced with inducible Fas or FADD undergo apoptosis after exposure to CID.
- T- Vehicles altered with the therapeutic attribute of loaded and/or engineered to in- vivo imaging Positron emission tomography (PET) is a nuclear medicine imaging technique that produces a three-dimensional image or picture of functional processes in the body. The system detects pairs of gamma rays emitted indirectly by a positron-emitting radionuclide (tracer), which is introduced into the body on a biologically active molecule. Three-dimensional images of tracer concentration within the body are then constructed by computer analysis. Due to the ability of the T-Vehicle to migrate to the tumor site, T-vehicles can be loaded with radioisotopes to allow the in-vivo detection and determine the location of a tumor site.
- PET Positron emission tomography
- Iodine- 123 (1231 or 1-123) is a radioactive isotope of iodine used in nuclear medicine imaging, including single photon emission computed tomography (SPECT). This is the most suitable isotope for the diagnostic study of thyroid diseases. The half-life of approximately 13.3 h (hours) is ideal for the 24-h (hour) iodine uptake test and 1231 has other advantages for diagnostically imaging thyroid tissue and thyroid cancer metastasis.
- Iodine can be used in a safe manner to image, or treat the thyroid tumor, due to the selective capture of Iodine in the "Iodine trap" by the hydrogen peroxide generated by the enzyme thyroid peroxidase (TPO). In this way, T-vehicles could be modified with Thyroid peroxidase or thyroperoxidase (TPO) to trap Iodine which can then be used to image/or kill the T- Vehicles.
- TPO thyroid peroxidase
- T-Vehicles can be created by many techniques including but not limited to any of the following:
- a) genetic modification with a viral vector such as retrovirus, adenovirus, Adeno-associated virus or lentivirus, and/or
- non-viral vectors including the use of DNA and/or R A vectors which are incorporated by physical and/or chemical techniques such as electroporation and/or lipofection methods using transposons and transposases (e.g. Sleeping Beauty), and/or Piggybac techniques, and/or
- transgenes that modify T-Vehicle migration, incorporate a suicide gene, improve recipient immune reconstitution (e.g. cytokine production), and/or elicit a direct anti-viral or anti-tumor effect (e.g. chimeric antigen receptor) or suppress the immune response for the treatment of auto immune diseases, and/or
- chemokine receptors such as CCR1, CCR2, CCR3, CCR4, CCR5, CCR6, CCR7, CCR8, CCR9, CCR10, CXCR1 , CXCR2, CXCR3-A, CXCR5, CXCR6, CX 3 CR1, and/or XCR1 to improve the migration of the T-Vehicle, and/or
- cytokines such as GM-CSF, TNFa, INFy, IL2, IL8 IL15, IL7, IL12, IL21 or IL26 or through the expression or over expression of co-stimulatory molecules CD80,CD86, 41BBL, OX40L, and/or
- T-Vehicle death by the expression of one or more suicide genes such as thymidine kinase TK gene, CD20, CD 19 or iCaspase9, and/or
- transgenes such as chimeric antigen receptors (CARs) that recognize tumor targets through single-chain variable fragments (scFv) isolated from specific antibodies linked with i) an extracellular spacer such as by the use of the CH2CH3 sequence derived from the IgG-FC region, or ii) a trans-membrane component including but not limited to the sequence of CD28, CD4, CD3 or CD8, iii) CD3 ⁇ endodomain or iv) by the expression of natural ligands such as cytokines or cytokines receptors encoding the CD3 ⁇ endodomain, and/or
- CARs chimeric antigen receptors
- scFv single-chain variable fragments
- transgenes that produce one or more immunosuppressive cytokines such as IL4, IL6, IL10, IL13, TFGP, or by the expression of competitor ligands such as CTLA-4, PD1.
- immunosuppressive cytokines such as IL4, IL6, IL10, IL13, TFGP
- competitor ligands such as CTLA-4, PD1.
- T- Vehicles can be wide ranging including but not limited to any of the following:
- c) as a biological vehicle allows to carry chemical compound with therapeutic purpose including but not limited to chemotherapy drugs, small molecules, nanoparticles, hormonal agonist or antagonist, anti-viral, anti-fungal, anti-parasitic agent, and/or d) as a biological vehicle to carry chemical compound(s) with no therapeutic purpose but secondary gain including but not limited to in-vivo identification and imaging that will allow to identify metastatic disease sites.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Immunology (AREA)
- Chemical & Material Sciences (AREA)
- General Health & Medical Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Cell Biology (AREA)
- Medicinal Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Epidemiology (AREA)
- Microbiology (AREA)
- Mycology (AREA)
- Engineering & Computer Science (AREA)
- Organic Chemistry (AREA)
- Biomedical Technology (AREA)
- Zoology (AREA)
- Biotechnology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Genetics & Genomics (AREA)
- Developmental Biology & Embryology (AREA)
- Virology (AREA)
- Wood Science & Technology (AREA)
- General Chemical & Material Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Hematology (AREA)
- Biochemistry (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Oncology (AREA)
- Reproductive Health (AREA)
- General Engineering & Computer Science (AREA)
- Hospice & Palliative Care (AREA)
- Toxicology (AREA)
- Gastroenterology & Hepatology (AREA)
- Biophysics (AREA)
- Molecular Biology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
- Medicines Containing Material From Animals Or Micro-Organisms (AREA)
Priority Applications (12)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
JP2015517361A JP2015519080A (ja) | 2012-06-11 | 2013-06-11 | 養子細胞療法のための改良型細胞培養方法 |
IL302514A IL302514A (en) | 2012-06-11 | 2013-06-11 | Improved cell culture methods for stress cell therapy |
CN201380030610.XA CN104411819B (zh) | 2012-06-11 | 2013-06-11 | 用于过继细胞疗法的改进的细胞培养方法 |
IL288241A IL288241B2 (en) | 2012-06-11 | 2013-06-11 | Improved cell culture methods for stress cell therapy |
SG11201407819UA SG11201407819UA (en) | 2012-06-11 | 2013-06-11 | Improved methods of cell culture for adoptive cell therapy |
EP13804099.3A EP2859093A4 (en) | 2012-06-11 | 2013-06-11 | IMPROVED METHODS FOR CELL CULTURES FOR ADOPTIVE CELL THERAPIES |
AU2013274416A AU2013274416B2 (en) | 2012-06-11 | 2013-06-11 | Improved methods of cell culture for adoptive cell therapy |
CA2873608A CA2873608A1 (en) | 2012-06-11 | 2013-06-11 | Improved methods of cell culture for adoptive cell therapy |
IL235739A IL235739B (en) | 2012-06-11 | 2014-11-17 | Improved cell culture methods for stress cell therapy |
AU2019240684A AU2019240684A1 (en) | 2012-06-11 | 2019-10-03 | Improved methods of cell culture for adoptive cell therapy |
IL273719A IL273719B (en) | 2012-06-11 | 2020-03-31 | Improved cell culture methods for stress cell therapy |
AU2022202172A AU2022202172A1 (en) | 2012-06-11 | 2022-03-30 | Improved methods of cell culture for adoptive cell therapy |
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US13/493,768 | 2012-06-11 | ||
US13/493,768 US20130115617A1 (en) | 2009-12-08 | 2012-06-11 | Methods of cell culture for adoptive cell therapy |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2013188427A1 true WO2013188427A1 (en) | 2013-12-19 |
Family
ID=49758673
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2013/045209 WO2013188427A1 (en) | 2012-06-11 | 2013-06-11 | Improved methods of cell culture for adoptive cell therapy |
Country Status (8)
Country | Link |
---|---|
EP (1) | EP2859093A4 (zh) |
JP (4) | JP2015519080A (zh) |
CN (2) | CN110241086A (zh) |
AU (3) | AU2013274416B2 (zh) |
CA (1) | CA2873608A1 (zh) |
IL (4) | IL302514A (zh) |
SG (2) | SG11201407819UA (zh) |
WO (1) | WO2013188427A1 (zh) |
Cited By (55)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN106795497A (zh) * | 2014-08-12 | 2017-05-31 | 人类起源公司 | 被工程化以归巢至淋巴结b细胞区、皮肤或胃肠道的car‑t淋巴细胞 |
WO2018081473A1 (en) | 2016-10-26 | 2018-05-03 | Iovance Biotherapeutics, Inc. | Restimulation of cryopreserved tumor infiltrating lymphocytes |
WO2018081789A1 (en) | 2016-10-31 | 2018-05-03 | Iovance Biotherapeutics, Inc. | Engineered artificial antigen presenting cells for tumor infiltrating lymphocyte expansion |
WO2018094167A1 (en) | 2016-11-17 | 2018-05-24 | Iovance Biotherapeutics, Inc. | Remnant tumor infiltrating lymphocytes and methods of preparing and using the same |
WO2018129332A1 (en) | 2017-01-06 | 2018-07-12 | Iovance Biotherapeutics, Inc. | Expansion of tumor infiltrating lymphocytes (tils) with tumor necrosis factor receptor superfamily (tnfrsf) agonists and therapeutic combinations of tils and tnfrsf agonists |
WO2018129336A1 (en) | 2017-01-06 | 2018-07-12 | Iovance Biotherapeutics, Inc. | Expansion of tumor infiltrating lymphocytes with potassium channel agonists and therapeutic uses thereof |
WO2018182817A1 (en) | 2017-03-29 | 2018-10-04 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
WO2018209115A1 (en) | 2017-05-10 | 2018-11-15 | Iovance Biotherapeutics, Inc. | Expansion of tumor infiltrating lymphocytes from liquid tumors and therapeutic uses thereof |
WO2018226714A1 (en) | 2017-06-05 | 2018-12-13 | Iovance Biotherapeutics, Inc. | Methods of using tumor infiltrating lymphocytes in double-refractory melanoma |
WO2019100023A1 (en) | 2017-11-17 | 2019-05-23 | Iovance Biotherapeutics, Inc. | Til expansion from fine needle aspirates and small biopsies |
WO2019103857A1 (en) | 2017-11-22 | 2019-05-31 | Iovance Biotherapeutics, Inc. | Expansion of peripheral blood lymphocytes (pbls) from peripheral blood |
WO2019118873A2 (en) | 2017-12-15 | 2019-06-20 | Iovance Biotherapeutics, Inc. | Systems and methods for determining the beneficial administration of tumor infiltrating lymphocytes, and methods of use thereof and beneficial administration of tumor infiltrating lymphocytes, and methods of use thereof |
WO2019136459A1 (en) | 2018-01-08 | 2019-07-11 | Iovance Biotherapeutics, Inc. | Processes for generating til products enriched for tumor antigen-specific t-cells |
WO2019136456A1 (en) | 2018-01-08 | 2019-07-11 | Iovance Biotherapeutics, Inc. | Processes for generating til products enriched for tumor antigen-specific t-cells |
WO2019190579A1 (en) | 2018-03-29 | 2019-10-03 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
WO2019210131A1 (en) | 2018-04-27 | 2019-10-31 | Iovance Biotherapeutics, Inc. | Closed process for expansion and gene editing of tumor infiltrating lymphocytes and uses of same in immunotherapy |
WO2019217753A1 (en) | 2018-05-10 | 2019-11-14 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
US10533156B2 (en) | 2009-12-08 | 2020-01-14 | Baylor College Of Medicine | Methods of cell culture for adoptive cell therapy |
WO2020061429A1 (en) | 2018-09-20 | 2020-03-26 | Iovance Biotherapeutics, Inc. | Expansion of tils from cryopreserved tumor samples |
WO2020096989A1 (en) | 2018-11-05 | 2020-05-14 | Iovance Biotherapeutics, Inc. | Treatment of nsclc patients refractory for anti-pd-1 antibody |
WO2020096927A1 (en) | 2018-11-05 | 2020-05-14 | Iovance Biotherapeutics, Inc. | Expansion of tils utilizing akt pathway inhibitors |
WO2020096682A2 (en) | 2018-08-31 | 2020-05-14 | Iovance Biotherapeutics, Inc. | Treatment of nsclc patients refractory for anti-pd-1 antibody |
WO2020096986A2 (en) | 2018-11-05 | 2020-05-14 | Iovance Biotherapeutics, Inc. | Selection of improved tumor reactive t-cells |
WO2020096988A2 (en) | 2018-11-05 | 2020-05-14 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of the same in immunotherapy |
WO2020131547A1 (en) | 2018-12-19 | 2020-06-25 | Iovance Biotherapeutics, Inc. | Methods of expanding tumor infiltrating lymphocytes using engineered cytokine receptor pairs and uses thereof |
WO2020232029A1 (en) | 2019-05-13 | 2020-11-19 | Iovance Biotherapeutics, Inc. | Methods and compositions for selecting tumor infiltrating lymphocytes and uses of the same in immunotherapy |
US10967005B2 (en) | 2013-03-15 | 2021-04-06 | Celgene Corporation | Modified T lymphocytes comprising a BAFF antibody-inducible caspase and methods of apoptosis |
WO2021081378A1 (en) | 2019-10-25 | 2021-04-29 | Iovance Biotherapeutics, Inc. | Gene editing of tumor infiltrating lymphocytes and uses of same in immunotherapy |
WO2021118990A1 (en) | 2019-12-11 | 2021-06-17 | Iovance Biotherapeutics, Inc. | Processes for the production of tumor infiltrating lymphocytes (tils) and methods of using the same |
US11130820B2 (en) | 2012-12-20 | 2021-09-28 | Celgene Corporation | Chimeric antigen receptors |
US11141434B2 (en) | 2016-07-07 | 2021-10-12 | Iovance Biotherapeutics, Inc. | Programmed death 1 ligand 1 (PD-L1) binding proteins and methods of use thereof |
WO2021216920A1 (en) | 2020-04-22 | 2021-10-28 | Iovance Biotherapeutics, Inc. | Systems and methods for coordinating manufacturing of cells for patient-specific immunotherapy |
WO2021226061A1 (en) | 2020-05-04 | 2021-11-11 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of the same in immunotherapy |
WO2021226085A1 (en) | 2020-05-04 | 2021-11-11 | Iovance Biotherapeutics, Inc. | Selection of improved tumor reactive t-cells |
US11254913B1 (en) | 2017-03-29 | 2022-02-22 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
WO2022076952A1 (en) | 2020-10-06 | 2022-04-14 | Iovance Biotherapeutics, Inc. | Treatment of nsclc patients with tumor infiltrating lymphocyte therapies |
WO2022076606A1 (en) | 2020-10-06 | 2022-04-14 | Iovance Biotherapeutics, Inc. | Treatment of nsclc patients with tumor infiltrating lymphocyte therapies |
WO2022125941A1 (en) | 2020-12-11 | 2022-06-16 | Iovance Biotherapeutics, Inc. | Treatment of cancer patients with tumor infiltrating lymphocyte therapies in combination with braf inhibitors and/or mek inhibitors |
WO2022133140A1 (en) | 2020-12-17 | 2022-06-23 | Iovance Biotherapeutics, Inc. | Treatment with tumor infiltrating lymphocyte therapies in combination with ctla-4 and pd-1 inhibitors |
WO2022133149A1 (en) | 2020-12-17 | 2022-06-23 | Iovance Biotherapeutics, Inc. | Treatment of cancers with tumor infiltrating lymphocytes |
US11401506B2 (en) | 2014-04-10 | 2022-08-02 | H. Lee Moffitt Cancer Center And Research Institute, Inc. | Enhanced expansion of tumor-infiltrating lymphocytes for adoptive cell therapy |
WO2022225981A2 (en) | 2021-04-19 | 2022-10-27 | Iovance Biotherapeutics, Inc. | Chimeric costimulatory receptors, chemokine receptors, and the use of same in cellular immunotherapies |
US11530386B2 (en) | 2015-12-15 | 2022-12-20 | Instil Bio (Uk) Limited | Cells expressing recombinant growth factor receptors |
WO2023009716A1 (en) | 2021-07-28 | 2023-02-02 | Iovance Biotherapeutics, Inc. | Treatment of cancer patients with tumor infiltrating lymphocyte therapies in combination with kras inhibitors |
US11618878B2 (en) | 2017-01-13 | 2023-04-04 | Instil Bio (Uk) Limited | Aseptic tissue processing method, kit and device |
WO2023077015A2 (en) | 2021-10-27 | 2023-05-04 | Iovance Biotherapeutics, Inc. | Systems and methods for coordinating manufacturing of cells for patient-specific immunotherapy |
WO2023086803A1 (en) | 2021-11-10 | 2023-05-19 | Iovance Biotherapeutics, Inc. | Methods of expansion treatment utilizing cd8 tumor infiltrating lymphocytes |
US11713446B2 (en) | 2018-01-08 | 2023-08-01 | Iovance Biotherapeutics, Inc. | Processes for generating TIL products enriched for tumor antigen-specific T-cells |
WO2023147486A1 (en) | 2022-01-28 | 2023-08-03 | Iovance Biotherapeutics, Inc. | Tumor infiltrating lymphocytes engineered to express payloads |
US11767510B2 (en) | 2019-12-20 | 2023-09-26 | Instil Bio (Uk) Limited | Devices and methods for isolating tumor infiltrating lymphocytes and uses thereof |
WO2024030758A1 (en) | 2022-08-01 | 2024-02-08 | Iovance Biotherapeutics, Inc. | Chimeric costimulatory receptors, chemokine receptors, and the use of same in cellular immunotherapies |
US11981921B2 (en) | 2022-04-15 | 2024-05-14 | Iovance Biotherapeutics, Inc. | TIL expansion processes using specific cytokine combinations and/or AKTi treatment |
WO2024118836A1 (en) | 2022-11-30 | 2024-06-06 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes with shortened rep step |
US12006354B2 (en) | 2017-05-24 | 2024-06-11 | Novartis Ag | Antibody-IL2 engrafted proteins and methods of use in the treatment of cancer |
WO2024151885A1 (en) | 2023-01-13 | 2024-07-18 | Iovance Biotherapeutics, Inc. | Use of til as maintenance therapy for nsclc patients who achieved pr/cr after prior therapy |
Families Citing this family (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
JP2015519080A (ja) * | 2012-06-11 | 2015-07-09 | ウィルソン ウォルフ マニュファクチャリング コーポレイションWilson Wolf Manufacturing Corporation | 養子細胞療法のための改良型細胞培養方法 |
CN106755023A (zh) * | 2015-10-15 | 2017-05-31 | 中国人民解放军军事医学科学院附属医院 | 带安全开关的嵌合抗原受体免疫细胞及其制备方法与应用 |
CN106119193B (zh) * | 2016-07-28 | 2019-10-29 | 上海闪锦生物科技有限公司 | 一种兼有nk细胞特质的抗原特异性t细胞的制备方法 |
Citations (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US5731160A (en) * | 1992-05-26 | 1998-03-24 | Rijksuniversiteit Leiden | Induction of antigen specific T-lymphocyte responses by stimulation with peptide loaded MHC class I molecules on antigen processing defective mammalian cell lines |
US20030235908A1 (en) * | 2000-02-24 | 2003-12-25 | Xcyte Therapies, Inc. | Activation and expansion of cells |
US20100261269A1 (en) * | 2002-01-03 | 2010-10-14 | The Trustees Of The University Of Pennsylvania | Activation and Expansion of T-Cells Using An Engineered Multivalent Signaling Platform as a Research Tool |
US20110136228A1 (en) * | 2009-12-08 | 2011-06-09 | Vera Juan F | Methods of cell culture for adoptive cell therapy |
Family Cites Families (22)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US7670781B2 (en) * | 2002-01-03 | 2010-03-02 | The Trustees Of The University Of Pennsylvania | Activation and expansion of T-cells using an agent that provides a primary activation signal and another agent that provides a co-stimulatory signal |
AU2003300359A1 (en) * | 2003-05-08 | 2004-12-13 | Xcyte Therapies, Inc. | Generation and isolation of antigen-specific t cells |
DK2327763T3 (en) * | 2005-08-05 | 2018-05-22 | Helmholtz Zentrum Muenchen Deutsches Forschungszentrum Gesundheit & Umwelt Gmbh | Antigen generation of specific T cells |
JP2009213462A (ja) * | 2008-02-15 | 2009-09-24 | Kist-Europe Forschungs Gmbh | 細胞改変方法および細胞改変装置 |
EP3854870A1 (en) * | 2009-08-24 | 2021-07-28 | Baylor College of Medicine | Generation of ctl lines with specificity against multiple tumor antigens or multiple viruses |
US20130115617A1 (en) * | 2009-12-08 | 2013-05-09 | John R. Wilson | Methods of cell culture for adoptive cell therapy |
US10548921B2 (en) * | 2011-04-08 | 2020-02-04 | Baylor College Of Medicine | Reversing the effects of the tumor microenvironment using chimeric cytokine receptors |
AU2012305931B2 (en) * | 2011-09-08 | 2017-09-07 | Yeda Research And Development Co. Ltd | Anti third party central memory T cells, methods of producing same and use of same in transplantation and disease treatment |
GB201121308D0 (en) * | 2011-12-12 | 2012-01-25 | Cell Medica Ltd | Process |
CN104769104A (zh) * | 2011-12-12 | 2015-07-08 | 细胞药物有限公司 | 扩大t细胞的方法 |
EP4089167A1 (en) * | 2012-02-09 | 2022-11-16 | Baylor College of Medicine | Pepmixes to generate multiviral ctls with broad specificity |
US20130217122A1 (en) * | 2012-02-21 | 2013-08-22 | The Trustees Of The University Of Pennsylvania | Expansion of Interferon-Gamma-Producing T-Cells Using Glypican-3 Peptide Library |
CN102719399A (zh) * | 2012-04-28 | 2012-10-10 | 北京爱根生物科技有限公司 | 自体特异性t细胞的体外扩增方法及所制备的t细胞体系、体系的药物应用和组分监测方法 |
SG11201407226WA (en) * | 2012-05-18 | 2014-12-30 | Wolf Wilson Mfg Corp | Improved methods of cell culture for adoptive cell therapy |
JP2015519080A (ja) * | 2012-06-11 | 2015-07-09 | ウィルソン ウォルフ マニュファクチャリング コーポレイションWilson Wolf Manufacturing Corporation | 養子細胞療法のための改良型細胞培養方法 |
IL290429B2 (en) * | 2013-09-23 | 2024-02-01 | Wilson Wolf Mfg Corporation | Improved methods for genetic modification in animal cells |
EP3515461A1 (en) * | 2016-09-23 | 2019-07-31 | Memorial Sloan Kettering Cancer Center | Generation and use in adoptive immunotherapy of stem cell-like memory t cells |
EP3516043A1 (en) * | 2016-09-26 | 2019-07-31 | Tessa Therapeutics Pte. Ltd. | T cell expansion method |
JP2020527044A (ja) * | 2017-07-13 | 2020-09-03 | アンセルム(アンスティチュート・ナシオナル・ドゥ・ラ・サンテ・エ・ドゥ・ラ・ルシェルシュ・メディカル) | CD8+CD45RCLOW/−Treg集団の拡大増殖および免疫抑制能力を高めるための方法 |
WO2021021937A1 (en) * | 2019-07-29 | 2021-02-04 | Baylor College Of Medicine | Antigen-specific t cell banks and methods of making and using the same therapeutically |
EP4013897A4 (en) * | 2019-08-16 | 2024-07-24 | Baylor College Medicine | T-CELL COMPOSITIONS SPECIFIC FOR A THIRD PARTY VIRUS, AND METHODS OF MANUFACTURE AND USE THEREOF IN ANTIVIRAL PROPHYLAXIS |
US20210371822A1 (en) * | 2020-05-27 | 2021-12-02 | University Of Southern California | Methods for expanding sars-cov2-antigen-specific t cells, compositions and uses related thereto |
-
2013
- 2013-06-11 JP JP2015517361A patent/JP2015519080A/ja active Pending
- 2013-06-11 CN CN201910301618.4A patent/CN110241086A/zh active Pending
- 2013-06-11 CN CN201380030610.XA patent/CN104411819B/zh active Active
- 2013-06-11 CA CA2873608A patent/CA2873608A1/en active Pending
- 2013-06-11 WO PCT/US2013/045209 patent/WO2013188427A1/en active Application Filing
- 2013-06-11 AU AU2013274416A patent/AU2013274416B2/en active Active
- 2013-06-11 SG SG11201407819UA patent/SG11201407819UA/en unknown
- 2013-06-11 IL IL302514A patent/IL302514A/en unknown
- 2013-06-11 IL IL288241A patent/IL288241B2/en unknown
- 2013-06-11 SG SG10201610387QA patent/SG10201610387QA/en unknown
- 2013-06-11 EP EP13804099.3A patent/EP2859093A4/en active Pending
-
2014
- 2014-11-17 IL IL235739A patent/IL235739B/en active IP Right Grant
-
2018
- 2018-06-15 JP JP2018114352A patent/JP2018138059A/ja active Pending
-
2019
- 2019-10-03 AU AU2019240684A patent/AU2019240684A1/en not_active Abandoned
-
2020
- 2020-03-31 IL IL273719A patent/IL273719B/en unknown
- 2020-08-07 JP JP2020134448A patent/JP7244461B2/ja active Active
-
2022
- 2022-03-30 AU AU2022202172A patent/AU2022202172A1/en active Pending
-
2023
- 2023-03-08 JP JP2023035371A patent/JP2023065668A/ja active Pending
Patent Citations (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US5731160A (en) * | 1992-05-26 | 1998-03-24 | Rijksuniversiteit Leiden | Induction of antigen specific T-lymphocyte responses by stimulation with peptide loaded MHC class I molecules on antigen processing defective mammalian cell lines |
US20030235908A1 (en) * | 2000-02-24 | 2003-12-25 | Xcyte Therapies, Inc. | Activation and expansion of cells |
US20100261269A1 (en) * | 2002-01-03 | 2010-10-14 | The Trustees Of The University Of Pennsylvania | Activation and Expansion of T-Cells Using An Engineered Multivalent Signaling Platform as a Research Tool |
US20110136228A1 (en) * | 2009-12-08 | 2011-06-09 | Vera Juan F | Methods of cell culture for adoptive cell therapy |
Non-Patent Citations (1)
Title |
---|
ULRIKE, GERDEMANN ET AL.: "Generation of multivirus-specific T cells to prevent/treat viral infections after allogeneic hematopoietic stem cell transplant", JOURNAL OF VISUALIZED EXPERIMENTS, vol. 51, 27 May 2011 (2011-05-27), pages 1 - 6, XP055034564 * |
Cited By (150)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US11268066B2 (en) | 2009-12-08 | 2022-03-08 | Wilson Wolf Manufacturing | Methods of cell culture for adoptive cell therapy |
US11999969B2 (en) | 2009-12-08 | 2024-06-04 | Wilson Wolf Manufacturing | Methods of cell culture for adoptive cell therapy |
US10533156B2 (en) | 2009-12-08 | 2020-01-14 | Baylor College Of Medicine | Methods of cell culture for adoptive cell therapy |
US11130820B2 (en) | 2012-12-20 | 2021-09-28 | Celgene Corporation | Chimeric antigen receptors |
US11806365B2 (en) | 2013-03-15 | 2023-11-07 | Celgene Corporation | Modified T lymphocytes comprising a CD52 antibody-inducible caspase and methods of apoptosis |
US10967005B2 (en) | 2013-03-15 | 2021-04-06 | Celgene Corporation | Modified T lymphocytes comprising a BAFF antibody-inducible caspase and methods of apoptosis |
US11401506B2 (en) | 2014-04-10 | 2022-08-02 | H. Lee Moffitt Cancer Center And Research Institute, Inc. | Enhanced expansion of tumor-infiltrating lymphocytes for adoptive cell therapy |
EP3828267A3 (en) * | 2014-08-12 | 2021-08-11 | Celgene Corporation | Car-t lymphocytes engineered to home to lymph node b cell zone, skin, or gastrointestinal tract |
CN106795497A (zh) * | 2014-08-12 | 2017-05-31 | 人类起源公司 | 被工程化以归巢至淋巴结b细胞区、皮肤或胃肠道的car‑t淋巴细胞 |
EP3194579A4 (en) * | 2014-08-12 | 2018-04-04 | Anthrogenesis Corporation | Car-t lymphocytes engineered to home to lymph node b cell zone, skin, or gastrointestinal tract |
US11530386B2 (en) | 2015-12-15 | 2022-12-20 | Instil Bio (Uk) Limited | Cells expressing recombinant growth factor receptors |
US12023355B2 (en) | 2016-07-07 | 2024-07-02 | Iovance Biotherapeutics, Inc. | Programmed death 1 ligand 1 (PD-L1) binding proteins and methods of use thereof |
US11141434B2 (en) | 2016-07-07 | 2021-10-12 | Iovance Biotherapeutics, Inc. | Programmed death 1 ligand 1 (PD-L1) binding proteins and methods of use thereof |
US11311578B2 (en) | 2016-10-26 | 2022-04-26 | Iovance Biotherapeutics, Inc. | Restimulation of cryopreserved tumor infiltrating lymphocytes |
US11058728B1 (en) | 2016-10-26 | 2021-07-13 | Iovance Biotherapeutics, Inc. | Restimulation of cryopreserved tumor infiltrating lymphocytes |
US11364266B2 (en) | 2016-10-26 | 2022-06-21 | Iovance Biotherapeutics, Inc. | Restimulation of cryopreserved tumor infiltrating lymphocytes |
US11266694B2 (en) | 2016-10-26 | 2022-03-08 | Iovance Biotherapeutics, Inc. | Restimulation of cryopreserved tumor infiltrating lymphocytes |
US11975028B2 (en) | 2016-10-26 | 2024-05-07 | Iovance Biotherapeutics, Inc. | Restimulation of cryopreserved tumor infiltrating lymphocytes |
US11179419B2 (en) | 2016-10-26 | 2021-11-23 | Iovance Biotherapeutics, Inc. | Restimulation of cryopreserved tumor infiltrating lymphocytes |
US11304980B2 (en) | 2016-10-26 | 2022-04-19 | Iovance Biotherapeutics, Inc. | Restimulation of cryopreserved tumor infiltrating lymphocytes |
WO2018081473A1 (en) | 2016-10-26 | 2018-05-03 | Iovance Biotherapeutics, Inc. | Restimulation of cryopreserved tumor infiltrating lymphocytes |
US11344580B2 (en) | 2016-10-26 | 2022-05-31 | Iovance Biotherapeutics, Inc. | Restimulation of cryopreserved tumor infiltrating lymphocytes |
US11969444B2 (en) | 2016-10-26 | 2024-04-30 | Iovance Biotherapeutics, Inc. | Restimulation of cryopreserved tumor infiltrating lymphocytes |
US11141438B2 (en) | 2016-10-26 | 2021-10-12 | Iovance Biotherapeutics, Inc. | Restimulation of cryopreserved tumor infiltrating lymphocytes |
US11344581B2 (en) | 2016-10-26 | 2022-05-31 | Iovance Biotherapeutics, Inc. | Restimulation of cryopreserved tumor infiltrating lymphocytes |
US11865140B2 (en) | 2016-10-26 | 2024-01-09 | Iovance Biotherapeutics, Inc. | Restimulation of cryopreserved tumor infiltrating lymphocytes |
US10517894B2 (en) | 2016-10-26 | 2019-12-31 | Iovance Biotherapeutics, Inc. | Restimulation of cryopreserved tumor infiltrating lymphocytes |
US11351199B2 (en) | 2016-10-26 | 2022-06-07 | Iovance Biotherapeutics, Inc. | Restimulation of cryopreserved tumor infiltrating lymphocytes |
US11351198B2 (en) | 2016-10-26 | 2022-06-07 | Iovance Biotherapeutics, Inc. | Restimulation of cryopreserved tumor infiltrating lymphocytes |
US11857573B2 (en) | 2016-10-26 | 2024-01-02 | Iovance Biotherapeutics, Inc. | Restimulation of cryopreserved tumor infiltrating lymphocytes |
US11123371B2 (en) | 2016-10-26 | 2021-09-21 | Iovance Biotherapeutics, Inc. | Restimulation of cryopreserved tumor infiltrating lymphocytes |
US11351197B2 (en) | 2016-10-26 | 2022-06-07 | Iovante Biotherapeutics, Inc. | Restimulation of cryopreserved tumor infiltrating lymphocytes |
US11369637B2 (en) | 2016-10-26 | 2022-06-28 | Iovance Biotherapeutics, Inc. | Restimulation of cryopreserved tumor infiltrating lymphocytes |
US11026974B2 (en) | 2016-10-26 | 2021-06-08 | Iovance Biotherapeutics, Inc. | Restimulation of cryopreserved tumor infiltrating lymphocytes |
EP4180520A1 (en) | 2016-10-26 | 2023-05-17 | Iovance Biotherapeutics, Inc. | Restimulation of cryopreserved tumor infiltrating lymphocytes |
US11667890B2 (en) | 2016-10-31 | 2023-06-06 | Iovance Biotherapeutics, Inc. | Engineered artificial antigen presenting cells for tumor infiltrating lymphocyte expansion |
WO2018081789A1 (en) | 2016-10-31 | 2018-05-03 | Iovance Biotherapeutics, Inc. | Engineered artificial antigen presenting cells for tumor infiltrating lymphocyte expansion |
US10415015B2 (en) | 2016-10-31 | 2019-09-17 | Iovance Biotherapeutics, Inc. | Engineered artificial antigen presenting cells for tumor infiltrating lymphocyte expansion |
US11401507B2 (en) | 2016-11-17 | 2022-08-02 | Iovance Biotherapeutics, Inc. | Remnant tumor infiltrating lymphocytes and methods of preparing and using the same |
WO2018094167A1 (en) | 2016-11-17 | 2018-05-24 | Iovance Biotherapeutics, Inc. | Remnant tumor infiltrating lymphocytes and methods of preparing and using the same |
US11293009B2 (en) | 2016-11-17 | 2022-04-05 | Iovance Biotherapeutics, Inc. | Remnant tumor infiltrating lymphocytes and methods of preparing and using the same |
US11220670B2 (en) | 2016-11-17 | 2022-01-11 | Iovance Biotherapeutics, Inc. | Remnant tumor infiltrating lymphocytes and methods of preparing and using the same |
WO2018129332A1 (en) | 2017-01-06 | 2018-07-12 | Iovance Biotherapeutics, Inc. | Expansion of tumor infiltrating lymphocytes (tils) with tumor necrosis factor receptor superfamily (tnfrsf) agonists and therapeutic combinations of tils and tnfrsf agonists |
WO2018129336A1 (en) | 2017-01-06 | 2018-07-12 | Iovance Biotherapeutics, Inc. | Expansion of tumor infiltrating lymphocytes with potassium channel agonists and therapeutic uses thereof |
US11357841B2 (en) | 2017-01-06 | 2022-06-14 | Iovance Biotherapeutics, Inc. | Expansion of tumor infiltrating lymphocytes with potassium channel agonists and therapeutic uses thereof |
US11618878B2 (en) | 2017-01-13 | 2023-04-04 | Instil Bio (Uk) Limited | Aseptic tissue processing method, kit and device |
US11618877B2 (en) | 2017-01-13 | 2023-04-04 | Instil Bio (Uk) Limited | Aseptic tissue processing method, kit and device |
US10463697B2 (en) | 2017-03-29 | 2019-11-05 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
EP3766964A1 (en) | 2017-03-29 | 2021-01-20 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
US10933094B2 (en) | 2017-03-29 | 2021-03-02 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
US10946044B2 (en) | 2017-03-29 | 2021-03-16 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
US10946045B2 (en) | 2017-03-29 | 2021-03-16 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
US10953047B2 (en) | 2017-03-29 | 2021-03-23 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
US10953046B2 (en) | 2017-03-29 | 2021-03-23 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
US10918666B2 (en) | 2017-03-29 | 2021-02-16 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
US10653723B1 (en) | 2017-03-29 | 2020-05-19 | Iovance, Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of the same in immunotherapy |
US11007226B2 (en) | 2017-03-29 | 2021-05-18 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
US11007225B1 (en) | 2017-03-29 | 2021-05-18 | lovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
US11013770B1 (en) | 2017-03-29 | 2021-05-25 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
US10905718B2 (en) | 2017-03-29 | 2021-02-02 | lovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
US11517592B1 (en) | 2017-03-29 | 2022-12-06 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
US11040070B2 (en) | 2017-03-29 | 2021-06-22 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
US11052116B2 (en) | 2017-03-29 | 2021-07-06 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
US11052115B2 (en) | 2017-03-29 | 2021-07-06 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
EP4279574A2 (en) | 2017-03-29 | 2023-11-22 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
US11083752B2 (en) | 2017-03-29 | 2021-08-10 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
US10646517B2 (en) | 2017-03-29 | 2020-05-12 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
US10639330B2 (en) | 2017-03-29 | 2020-05-05 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
US10537595B2 (en) | 2017-03-29 | 2020-01-21 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
US11541077B2 (en) | 2017-03-29 | 2023-01-03 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
US11939596B2 (en) | 2017-03-29 | 2024-03-26 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
US10420799B2 (en) | 2017-03-29 | 2019-09-24 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
US11168303B2 (en) | 2017-03-29 | 2021-11-09 | Iovance Biotherapeutics | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
US11168304B2 (en) | 2017-03-29 | 2021-11-09 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
US10925900B2 (en) | 2017-03-29 | 2021-02-23 | lovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
US10398734B2 (en) | 2017-03-29 | 2019-09-03 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
EP3910055A1 (en) | 2017-03-29 | 2021-11-17 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
US10363273B2 (en) | 2017-03-29 | 2019-07-30 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
US11202804B2 (en) | 2017-03-29 | 2021-12-21 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
US11202803B1 (en) | 2017-03-29 | 2021-12-21 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
US10894063B2 (en) | 2017-03-29 | 2021-01-19 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
US11241456B2 (en) | 2017-03-29 | 2022-02-08 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
US11254913B1 (en) | 2017-03-29 | 2022-02-22 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
US11998568B2 (en) | 2017-03-29 | 2024-06-04 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
US10695372B2 (en) | 2017-03-29 | 2020-06-30 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
US11273180B2 (en) | 2017-03-29 | 2022-03-15 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
US11273181B2 (en) | 2017-03-29 | 2022-03-15 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
US11291687B2 (en) | 2017-03-29 | 2022-04-05 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
US11529372B1 (en) | 2017-03-29 | 2022-12-20 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
US10272113B2 (en) | 2017-03-29 | 2019-04-30 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
US10166257B2 (en) | 2017-03-29 | 2019-01-01 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
US12121541B2 (en) | 2017-03-29 | 2024-10-22 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
US11304979B2 (en) | 2017-03-29 | 2022-04-19 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
US10130659B2 (en) | 2017-03-29 | 2018-11-20 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
US11337998B2 (en) | 2017-03-29 | 2022-05-24 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
EP3722415A1 (en) | 2017-03-29 | 2020-10-14 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
WO2018182817A1 (en) | 2017-03-29 | 2018-10-04 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
US11344579B2 (en) | 2017-03-29 | 2022-05-31 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
EP3730608A1 (en) | 2017-03-29 | 2020-10-28 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
WO2018209115A1 (en) | 2017-05-10 | 2018-11-15 | Iovance Biotherapeutics, Inc. | Expansion of tumor infiltrating lymphocytes from liquid tumors and therapeutic uses thereof |
US12006354B2 (en) | 2017-05-24 | 2024-06-11 | Novartis Ag | Antibody-IL2 engrafted proteins and methods of use in the treatment of cancer |
US11433097B2 (en) | 2017-06-05 | 2022-09-06 | Iovance Biotherapeutics, Inc. | Methods of using tumor infiltrating lymphocytes in double-refractory melanoma |
WO2018226714A1 (en) | 2017-06-05 | 2018-12-13 | Iovance Biotherapeutics, Inc. | Methods of using tumor infiltrating lymphocytes in double-refractory melanoma |
WO2020117233A1 (en) | 2017-06-05 | 2020-06-11 | Iovance Biotherapeutics, Inc. | Methods of using tumor infiltrating lymphocytes in double-refractory melanoma |
US11819517B2 (en) | 2017-06-05 | 2023-11-21 | Iovance Biotherapeutics, Inc. | Methods of using tumor infiltrating lymphocytes in double-refractory melanoma |
WO2019100023A1 (en) | 2017-11-17 | 2019-05-23 | Iovance Biotherapeutics, Inc. | Til expansion from fine needle aspirates and small biopsies |
WO2019103857A1 (en) | 2017-11-22 | 2019-05-31 | Iovance Biotherapeutics, Inc. | Expansion of peripheral blood lymphocytes (pbls) from peripheral blood |
WO2019118873A2 (en) | 2017-12-15 | 2019-06-20 | Iovance Biotherapeutics, Inc. | Systems and methods for determining the beneficial administration of tumor infiltrating lymphocytes, and methods of use thereof and beneficial administration of tumor infiltrating lymphocytes, and methods of use thereof |
WO2019136456A1 (en) | 2018-01-08 | 2019-07-11 | Iovance Biotherapeutics, Inc. | Processes for generating til products enriched for tumor antigen-specific t-cells |
US12104172B2 (en) | 2018-01-08 | 2024-10-01 | Iovance Biotherapeutics, Inc. | Processes for generating TIL products enriched for tumor antigen-specific t-cells |
US11713446B2 (en) | 2018-01-08 | 2023-08-01 | Iovance Biotherapeutics, Inc. | Processes for generating TIL products enriched for tumor antigen-specific T-cells |
WO2019136459A1 (en) | 2018-01-08 | 2019-07-11 | Iovance Biotherapeutics, Inc. | Processes for generating til products enriched for tumor antigen-specific t-cells |
WO2019190579A1 (en) | 2018-03-29 | 2019-10-03 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
EP4386080A2 (en) | 2018-03-29 | 2024-06-19 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
US11866688B2 (en) | 2018-04-27 | 2024-01-09 | Iovance Biotherapeutics, Inc. | Closed process for expansion and gene editing of tumor infiltrating lymphocytes and uses of same in immunotherapy |
US12031157B2 (en) | 2018-04-27 | 2024-07-09 | Iovance Biotherapeutics, Inc. | Closed process for expansion and gene editing of tumor infiltrating lymphocytes and uses of same in immunotherapy |
WO2019210131A1 (en) | 2018-04-27 | 2019-10-31 | Iovance Biotherapeutics, Inc. | Closed process for expansion and gene editing of tumor infiltrating lymphocytes and uses of same in immunotherapy |
US12024718B2 (en) | 2018-04-27 | 2024-07-02 | Iovance Biotherapeutics, Inc. | Closed process for expansion and gene editing of tumor infiltrating lymphocytes and uses of same in immunotherapy |
US11384337B2 (en) | 2018-04-27 | 2022-07-12 | Iovance Biotherapeutics, Inc. | Closed process for expansion and gene editing of tumor infiltrating lymphocytes and uses of same in immunotherapy |
WO2019217753A1 (en) | 2018-05-10 | 2019-11-14 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy |
WO2020096682A2 (en) | 2018-08-31 | 2020-05-14 | Iovance Biotherapeutics, Inc. | Treatment of nsclc patients refractory for anti-pd-1 antibody |
EP4378530A2 (en) | 2018-08-31 | 2024-06-05 | Iovance Biotherapeutics, Inc. | Use of tumor infiltrating lymphocytes for treating nsclc patients refractory for anti-pd-1 antibody |
WO2020061429A1 (en) | 2018-09-20 | 2020-03-26 | Iovance Biotherapeutics, Inc. | Expansion of tils from cryopreserved tumor samples |
WO2020096988A2 (en) | 2018-11-05 | 2020-05-14 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of the same in immunotherapy |
WO2020096986A2 (en) | 2018-11-05 | 2020-05-14 | Iovance Biotherapeutics, Inc. | Selection of improved tumor reactive t-cells |
WO2020096989A1 (en) | 2018-11-05 | 2020-05-14 | Iovance Biotherapeutics, Inc. | Treatment of nsclc patients refractory for anti-pd-1 antibody |
WO2020096927A1 (en) | 2018-11-05 | 2020-05-14 | Iovance Biotherapeutics, Inc. | Expansion of tils utilizing akt pathway inhibitors |
WO2020131547A1 (en) | 2018-12-19 | 2020-06-25 | Iovance Biotherapeutics, Inc. | Methods of expanding tumor infiltrating lymphocytes using engineered cytokine receptor pairs and uses thereof |
WO2020232029A1 (en) | 2019-05-13 | 2020-11-19 | Iovance Biotherapeutics, Inc. | Methods and compositions for selecting tumor infiltrating lymphocytes and uses of the same in immunotherapy |
WO2021081378A1 (en) | 2019-10-25 | 2021-04-29 | Iovance Biotherapeutics, Inc. | Gene editing of tumor infiltrating lymphocytes and uses of same in immunotherapy |
WO2021118990A1 (en) | 2019-12-11 | 2021-06-17 | Iovance Biotherapeutics, Inc. | Processes for the production of tumor infiltrating lymphocytes (tils) and methods of using the same |
US11767510B2 (en) | 2019-12-20 | 2023-09-26 | Instil Bio (Uk) Limited | Devices and methods for isolating tumor infiltrating lymphocytes and uses thereof |
US11631483B2 (en) | 2020-04-22 | 2023-04-18 | Iovance Biotherapeutics, Inc. | Systems and methods for coordinating manufacturing of cells for patient-specific immunotherapy |
WO2021216920A1 (en) | 2020-04-22 | 2021-10-28 | Iovance Biotherapeutics, Inc. | Systems and methods for coordinating manufacturing of cells for patient-specific immunotherapy |
WO2021226085A1 (en) | 2020-05-04 | 2021-11-11 | Iovance Biotherapeutics, Inc. | Selection of improved tumor reactive t-cells |
WO2021226061A1 (en) | 2020-05-04 | 2021-11-11 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes and uses of the same in immunotherapy |
WO2022076952A1 (en) | 2020-10-06 | 2022-04-14 | Iovance Biotherapeutics, Inc. | Treatment of nsclc patients with tumor infiltrating lymphocyte therapies |
WO2022076606A1 (en) | 2020-10-06 | 2022-04-14 | Iovance Biotherapeutics, Inc. | Treatment of nsclc patients with tumor infiltrating lymphocyte therapies |
WO2022125941A1 (en) | 2020-12-11 | 2022-06-16 | Iovance Biotherapeutics, Inc. | Treatment of cancer patients with tumor infiltrating lymphocyte therapies in combination with braf inhibitors and/or mek inhibitors |
WO2022133140A1 (en) | 2020-12-17 | 2022-06-23 | Iovance Biotherapeutics, Inc. | Treatment with tumor infiltrating lymphocyte therapies in combination with ctla-4 and pd-1 inhibitors |
WO2022133149A1 (en) | 2020-12-17 | 2022-06-23 | Iovance Biotherapeutics, Inc. | Treatment of cancers with tumor infiltrating lymphocytes |
WO2022225981A2 (en) | 2021-04-19 | 2022-10-27 | Iovance Biotherapeutics, Inc. | Chimeric costimulatory receptors, chemokine receptors, and the use of same in cellular immunotherapies |
WO2023009716A1 (en) | 2021-07-28 | 2023-02-02 | Iovance Biotherapeutics, Inc. | Treatment of cancer patients with tumor infiltrating lymphocyte therapies in combination with kras inhibitors |
WO2023077015A2 (en) | 2021-10-27 | 2023-05-04 | Iovance Biotherapeutics, Inc. | Systems and methods for coordinating manufacturing of cells for patient-specific immunotherapy |
WO2023086803A1 (en) | 2021-11-10 | 2023-05-19 | Iovance Biotherapeutics, Inc. | Methods of expansion treatment utilizing cd8 tumor infiltrating lymphocytes |
WO2023147486A1 (en) | 2022-01-28 | 2023-08-03 | Iovance Biotherapeutics, Inc. | Tumor infiltrating lymphocytes engineered to express payloads |
US11981921B2 (en) | 2022-04-15 | 2024-05-14 | Iovance Biotherapeutics, Inc. | TIL expansion processes using specific cytokine combinations and/or AKTi treatment |
WO2024030758A1 (en) | 2022-08-01 | 2024-02-08 | Iovance Biotherapeutics, Inc. | Chimeric costimulatory receptors, chemokine receptors, and the use of same in cellular immunotherapies |
WO2024118836A1 (en) | 2022-11-30 | 2024-06-06 | Iovance Biotherapeutics, Inc. | Processes for production of tumor infiltrating lymphocytes with shortened rep step |
WO2024151885A1 (en) | 2023-01-13 | 2024-07-18 | Iovance Biotherapeutics, Inc. | Use of til as maintenance therapy for nsclc patients who achieved pr/cr after prior therapy |
Also Published As
Publication number | Publication date |
---|---|
CA2873608A1 (en) | 2013-12-19 |
SG11201407819UA (en) | 2014-12-30 |
IL288241B2 (en) | 2023-10-01 |
SG10201610387QA (en) | 2017-02-27 |
JP7244461B2 (ja) | 2023-03-22 |
EP2859093A1 (en) | 2015-04-15 |
JP2020174685A (ja) | 2020-10-29 |
CN110241086A (zh) | 2019-09-17 |
JP2018138059A (ja) | 2018-09-06 |
IL273719B (en) | 2022-01-01 |
CN104411819A (zh) | 2015-03-11 |
JP2015519080A (ja) | 2015-07-09 |
CN104411819B (zh) | 2019-05-10 |
IL235739B (en) | 2020-04-30 |
AU2013274416A1 (en) | 2015-01-15 |
IL288241A (en) | 2022-01-01 |
AU2013274416B2 (en) | 2019-07-04 |
AU2022202172A1 (en) | 2022-04-21 |
AU2019240684A1 (en) | 2019-10-24 |
JP2023065668A (ja) | 2023-05-12 |
IL302514A (en) | 2023-07-01 |
IL288241B1 (en) | 2023-06-01 |
EP2859093A4 (en) | 2016-08-17 |
IL273719A (en) | 2020-05-31 |
IL235739A0 (en) | 2015-01-29 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
US20230383250A1 (en) | Methods of cell culture for adoptive cell therapy | |
JP7244461B2 (ja) | 養子細胞療法のための改良型細胞培養方法 | |
US11155784B2 (en) | Process of expanding T cells | |
US12060577B2 (en) | Compositions for expanding natural killer cells | |
Mahnke et al. | Maintenance of long‐term tumour‐specific T‐cell memory by residual dormant tumour cells | |
JP2015519080A5 (zh) | ||
Rooney et al. | T lymphocytes targeting native receptors | |
JP2017524031A (ja) | ガンマデルタt細胞およびその使用 | |
WO2009045308A2 (en) | Enhanced generation of cytotoxic t-lymphocytes by il-21 mediated foxp3 suppression | |
Durai et al. | In vivo functional efficacy of tumor-specific T cells expanded using HLA-Ig based artificial antigen presenting cells (aAPC) | |
Bollard et al. | Cytotoxic T lymphocytes for leukemia and lymphoma | |
Wang et al. | Stem cell like memory T cells: A new paradigm in cancer immunotherapy | |
SG187846A1 (en) | Cells expressing th1 characteristics and cytolytic properties | |
Li et al. | Artificial antigen-presenting cells: the booster for the obtaining of functional adoptive cells | |
CA2863557C (en) | Process of expanding t cells | |
Gerdemann et al. | Extending the use of adoptive T cell immunotherapy for infections and cancer | |
CN117979978A (zh) | 从干细胞产生经工程化的t细胞 | |
CN117940139A (zh) | 对用于同种异体car t细胞疗法的干细胞进行工程化 | |
CN118871572A (zh) | 使用多种t细胞受体对干细胞t细胞进行工程化 | |
Umeshappa et al. | CD4+ T CELLS PLAY CRITICAL ROLES VIA CD40L AND IL-2 SIGNALING IN MULTIPLE PHASES OF CD8+ CTL RESPONSES FOLLOWING ADENOVIRUS VACCINATION |
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: 13804099 Country of ref document: EP Kind code of ref document: A1 |
|
ENP | Entry into the national phase |
Ref document number: 2873608 Country of ref document: CA |
|
WWE | Wipo information: entry into national phase |
Ref document number: 235739 Country of ref document: IL |
|
ENP | Entry into the national phase |
Ref document number: 2015517361 Country of ref document: JP Kind code of ref document: A |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
WWE | Wipo information: entry into national phase |
Ref document number: 2013804099 Country of ref document: EP |
|
ENP | Entry into the national phase |
Ref document number: 2013274416 Country of ref document: AU Date of ref document: 20130611 Kind code of ref document: A |