NZ753978A - Binding polypeptides and methods of making the same - Google Patents
Binding polypeptides and methods of making the sameInfo
- Publication number
- NZ753978A NZ753978A NZ753978A NZ75397817A NZ753978A NZ 753978 A NZ753978 A NZ 753978A NZ 753978 A NZ753978 A NZ 753978A NZ 75397817 A NZ75397817 A NZ 75397817A NZ 753978 A NZ753978 A NZ 753978A
- Authority
- NZ
- New Zealand
- Prior art keywords
- cell
- strand
- cdna
- sequence
- chain
- Prior art date
Links
- 229920001184 polypeptide Polymers 0.000 title claims abstract description 166
- 230000027455 binding Effects 0.000 title claims description 153
- 102000004965 antibodies Human genes 0.000 claims abstract description 239
- 108090001123 antibodies Proteins 0.000 claims abstract description 239
- 210000004027 cells Anatomy 0.000 claims description 546
- 238000006243 chemical reaction Methods 0.000 claims description 332
- 229920002676 Complementary DNA Polymers 0.000 claims description 328
- 238000004519 manufacturing process Methods 0.000 claims description 307
- 239000002299 complementary DNA Substances 0.000 claims description 278
- 108091008153 T cell receptors Proteins 0.000 claims description 231
- 102000016266 T-Cell Antigen Receptors Human genes 0.000 claims description 229
- 150000007523 nucleic acids Chemical class 0.000 claims description 211
- 229920002106 messenger RNA Polymers 0.000 claims description 191
- 108020004999 Messenger RNA Proteins 0.000 claims description 190
- 239000000758 substrate Substances 0.000 claims description 150
- 125000003729 nucleotide group Chemical group 0.000 claims description 140
- 108020004707 nucleic acids Proteins 0.000 claims description 129
- 239000002773 nucleotide Substances 0.000 claims description 118
- 229920001850 Nucleic acid sequence Polymers 0.000 claims description 106
- 239000011324 bead Substances 0.000 claims description 106
- 239000000427 antigen Substances 0.000 claims description 103
- 102000038129 antigens Human genes 0.000 claims description 103
- 108091007172 antigens Proteins 0.000 claims description 103
- 230000000295 complement Effects 0.000 claims description 97
- 108090000364 Ligases Proteins 0.000 claims description 87
- 102000003960 Ligases Human genes 0.000 claims description 87
- 229920000272 Oligonucleotide Polymers 0.000 claims description 85
- 230000004048 modification Effects 0.000 claims description 80
- 238000006011 modification reaction Methods 0.000 claims description 80
- 238000003199 nucleic acid amplification method Methods 0.000 claims description 75
- 230000003321 amplification Effects 0.000 claims description 74
- 230000036647 reaction Effects 0.000 claims description 65
- 229920003013 deoxyribonucleic acid Polymers 0.000 claims description 60
- 230000000692 anti-sense Effects 0.000 claims description 40
- 239000003153 chemical reaction reagent Substances 0.000 claims description 40
- 239000000203 mixture Substances 0.000 claims description 40
- 101700043375 sing Proteins 0.000 claims description 38
- 238000000137 annealing Methods 0.000 claims description 33
- 230000002934 lysing Effects 0.000 claims description 31
- 230000015572 biosynthetic process Effects 0.000 claims description 29
- 230000009089 cytolysis Effects 0.000 claims description 26
- 239000002253 acid Substances 0.000 claims description 21
- 229920001223 polyethylene glycol Polymers 0.000 claims description 19
- 238000003786 synthesis reaction Methods 0.000 claims description 18
- 230000002194 synthesizing Effects 0.000 claims description 18
- PYMYPHUHKUWMLA-LMVFSUKVSA-N Ribose Natural products OC[C@@H](O)[C@@H](O)[C@@H](O)C=O PYMYPHUHKUWMLA-LMVFSUKVSA-N 0.000 claims description 14
- 238000003780 insertion Methods 0.000 claims description 5
- 229940035295 Ting Drugs 0.000 claims description 3
- 201000010099 disease Diseases 0.000 abstract description 44
- 239000000047 product Substances 0.000 description 81
- 241000282414 Homo sapiens Species 0.000 description 66
- 238000010839 reverse transcription Methods 0.000 description 58
- 238000002955 isolation Methods 0.000 description 54
- 235000001014 amino acid Nutrition 0.000 description 51
- 150000001413 amino acids Chemical class 0.000 description 50
- 229920000972 Sense strand Polymers 0.000 description 39
- 101700011961 DPOM Proteins 0.000 description 36
- 101710029649 MDV043 Proteins 0.000 description 36
- 101700061424 POLB Proteins 0.000 description 36
- 101700054624 RF1 Proteins 0.000 description 36
- 102000004190 Enzymes Human genes 0.000 description 32
- 108090000790 Enzymes Proteins 0.000 description 32
- 102000004169 proteins and genes Human genes 0.000 description 31
- 108090000623 proteins and genes Proteins 0.000 description 31
- 229940088598 Enzyme Drugs 0.000 description 27
- 235000018102 proteins Nutrition 0.000 description 27
- 239000000243 solution Substances 0.000 description 27
- 238000009396 hybridization Methods 0.000 description 24
- 239000010452 phosphate Substances 0.000 description 23
- 230000001225 therapeutic Effects 0.000 description 21
- 241000699666 Mus <mouse, genus> Species 0.000 description 20
- 239000000839 emulsion Substances 0.000 description 20
- 125000000539 amino acid group Chemical group 0.000 description 19
- 150000002500 ions Chemical class 0.000 description 19
- 229920000023 polynucleotide Polymers 0.000 description 19
- 239000002157 polynucleotide Substances 0.000 description 19
- 239000002202 Polyethylene glycol Substances 0.000 description 18
- 239000012071 phase Substances 0.000 description 18
- 201000009910 diseases by infectious agent Diseases 0.000 description 17
- 238000004166 bioassay Methods 0.000 description 16
- 239000003795 chemical substances by application Substances 0.000 description 15
- 108010045030 monoclonal antibodies Proteins 0.000 description 15
- 102000005614 monoclonal antibodies Human genes 0.000 description 15
- 102000018358 Immunoglobulins Human genes 0.000 description 14
- 108060003951 Immunoglobulins Proteins 0.000 description 14
- 241000700159 Rattus Species 0.000 description 14
- 240000004808 Saccharomyces cerevisiae Species 0.000 description 14
- 210000001744 T-Lymphocytes Anatomy 0.000 description 14
- ISAKRJDGNUQOIC-UHFFFAOYSA-N Uracil Chemical compound O=C1C=CNC(=O)N1 ISAKRJDGNUQOIC-UHFFFAOYSA-N 0.000 description 14
- 239000000463 material Substances 0.000 description 14
- 238000000034 method Methods 0.000 description 14
- 201000011510 cancer Diseases 0.000 description 13
- 230000001809 detectable Effects 0.000 description 13
- 239000000126 substance Substances 0.000 description 13
- 230000020382 suppression by virus of host antigen processing and presentation of peptide antigen via MHC class I Effects 0.000 description 13
- 206010028980 Neoplasm Diseases 0.000 description 12
- 125000003275 alpha amino acid group Chemical group 0.000 description 12
- UIIMBOGNXHQVGW-UHFFFAOYSA-M buffer Substances [Na+].OC([O-])=O UIIMBOGNXHQVGW-UHFFFAOYSA-M 0.000 description 12
- 230000001351 cycling Effects 0.000 description 12
- 230000000694 effects Effects 0.000 description 12
- 238000000338 in vitro Methods 0.000 description 12
- 239000000969 carrier Substances 0.000 description 11
- 238000005755 formation reaction Methods 0.000 description 11
- 101710030587 ligN Proteins 0.000 description 11
- 210000004962 mammalian cells Anatomy 0.000 description 11
- -1 nucleotide cation Chemical class 0.000 description 11
- 239000011541 reaction mixture Substances 0.000 description 11
- PJDOLCGOTSNFJM-UHFFFAOYSA-N 2,2,3,3,4,4,5,5,6,6,7,7,8,8,8-pentadecafluorooctan-1-ol Chemical compound OCC(F)(F)C(F)(F)C(F)(F)C(F)(F)C(F)(F)C(F)(F)C(F)(F)F PJDOLCGOTSNFJM-UHFFFAOYSA-N 0.000 description 10
- OKTJSMMVPCPJKN-UHFFFAOYSA-N carbon Chemical group [C] OKTJSMMVPCPJKN-UHFFFAOYSA-N 0.000 description 10
- 229910052799 carbon Inorganic materials 0.000 description 10
- 150000001875 compounds Chemical class 0.000 description 10
- 238000005538 encapsulation Methods 0.000 description 10
- DHMQDGOQFOQNFH-UHFFFAOYSA-N glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 description 10
- 230000002285 radioactive Effects 0.000 description 10
- 241000283984 Rodentia Species 0.000 description 9
- 238000004925 denaturation Methods 0.000 description 9
- 230000036425 denaturation Effects 0.000 description 9
- 239000003599 detergent Substances 0.000 description 9
- 238000002372 labelling Methods 0.000 description 9
- 101700077585 ligd Proteins 0.000 description 9
- 230000000051 modifying Effects 0.000 description 9
- 241000894007 species Species 0.000 description 9
- 108010092799 EC 2.7.7.49 Proteins 0.000 description 8
- 102000033147 ERVK-25 Human genes 0.000 description 8
- WUAPFZMCVAUBPE-UHFFFAOYSA-N Rhenium Chemical compound [Re] WUAPFZMCVAUBPE-UHFFFAOYSA-N 0.000 description 8
- NBIIXXVUZAFLBC-UHFFFAOYSA-K [O-]P([O-])([O-])=O Chemical compound [O-]P([O-])([O-])=O NBIIXXVUZAFLBC-UHFFFAOYSA-K 0.000 description 8
- QQINRWTZWGJFDB-UHFFFAOYSA-N actinium Chemical compound [Ac] QQINRWTZWGJFDB-UHFFFAOYSA-N 0.000 description 8
- YBJHBAHKTGYVGT-ZKWXMUAHSA-N biotin Chemical compound N1C(=O)N[C@@H]2[C@H](CCCCC(=O)O)SC[C@@H]21 YBJHBAHKTGYVGT-ZKWXMUAHSA-N 0.000 description 8
- 125000000267 glycino group Chemical group [H]N([*])C([H])([H])C(=O)O[H] 0.000 description 8
- 210000002865 immune cell Anatomy 0.000 description 8
- OHSVLFRHMCKCQY-UHFFFAOYSA-N lutetium Chemical compound [Lu] OHSVLFRHMCKCQY-UHFFFAOYSA-N 0.000 description 8
- 238000002823 phage display Methods 0.000 description 8
- OAICVXFJPJFONN-UHFFFAOYSA-N phosphorus Chemical compound [P] OAICVXFJPJFONN-UHFFFAOYSA-N 0.000 description 8
- 238000002264 polyacrylamide gel electrophoresis Methods 0.000 description 8
- YZCKVEUIGOORGS-NJFSPNSNSA-N tritium Chemical compound [3H] YZCKVEUIGOORGS-NJFSPNSNSA-N 0.000 description 8
- 230000035693 Fab Effects 0.000 description 7
- GSEJCLTVZPLZKY-UHFFFAOYSA-N Tris Chemical compound OCCN(CCO)CCO GSEJCLTVZPLZKY-UHFFFAOYSA-N 0.000 description 7
- 239000007983 Tris buffer Substances 0.000 description 7
- 229940035893 Uracil Drugs 0.000 description 7
- 238000010171 animal model Methods 0.000 description 7
- 229960002685 biotin Drugs 0.000 description 7
- 235000020958 biotin Nutrition 0.000 description 7
- 239000011616 biotin Substances 0.000 description 7
- 239000003814 drug Substances 0.000 description 7
- GNBHRKFJIUUOQI-UHFFFAOYSA-N fluorescein Chemical compound O1C(=O)C2=CC=CC=C2C21C1=CC=C(O)C=C1OC1=CC(O)=CC=C21 GNBHRKFJIUUOQI-UHFFFAOYSA-N 0.000 description 7
- 241000282836 Camelus dromedarius Species 0.000 description 6
- 241000283707 Capra Species 0.000 description 6
- 210000003169 Central Nervous System Anatomy 0.000 description 6
- AYFVYJQAPQTCCC-GBXIJSLDSA-N L-threonine Chemical compound C[C@@H](O)[C@H](N)C(O)=O AYFVYJQAPQTCCC-GBXIJSLDSA-N 0.000 description 6
- OUYCCCASQSFEME-QMMMGPOBSA-N L-tyrosine Chemical compound OC(=O)[C@@H](N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-QMMMGPOBSA-N 0.000 description 6
- 208000009874 Lice Infestations Diseases 0.000 description 6
- 241000283973 Oryctolagus cuniculus Species 0.000 description 6
- 231100000765 Toxin Toxicity 0.000 description 6
- 238000001990 intravenous administration Methods 0.000 description 6
- XEEYBQQBJWHFJM-UHFFFAOYSA-N iron Chemical compound [Fe] XEEYBQQBJWHFJM-UHFFFAOYSA-N 0.000 description 6
- 239000007788 liquid Substances 0.000 description 6
- 238000007481 next generation sequencing Methods 0.000 description 6
- 238000000159 protein binding assay Methods 0.000 description 6
- 230000002829 reduced Effects 0.000 description 6
- 230000004044 response Effects 0.000 description 6
- 239000003053 toxin Substances 0.000 description 6
- 230000035897 transcription Effects 0.000 description 6
- VWQVUPCCIRVNHF-UHFFFAOYSA-N yttrium Chemical compound [Y] VWQVUPCCIRVNHF-UHFFFAOYSA-N 0.000 description 6
- 206010049460 Abasia Diseases 0.000 description 5
- 108090001008 Avidin Proteins 0.000 description 5
- 210000003719 B-Lymphocytes Anatomy 0.000 description 5
- 229920000453 Consensus sequence Polymers 0.000 description 5
- 108010087819 Fc receptors Proteins 0.000 description 5
- 102000009109 Fc receptors Human genes 0.000 description 5
- 239000004471 Glycine Substances 0.000 description 5
- XUJNEKJLAYXESH-REOHCLBHSA-N L-cysteine Chemical compound SC[C@H](N)C(O)=O XUJNEKJLAYXESH-REOHCLBHSA-N 0.000 description 5
- AGPKZVBTJJNPAG-WHFBIAKZSA-N L-isoleucine Chemical compound CC[C@H](C)[C@H](N)C(O)=O AGPKZVBTJJNPAG-WHFBIAKZSA-N 0.000 description 5
- COLNVLDHVKWLRT-QMMMGPOBSA-N L-phenylalanine Chemical compound OC(=O)[C@@H](N)CC1=CC=CC=C1 COLNVLDHVKWLRT-QMMMGPOBSA-N 0.000 description 5
- MTCFGRXMJLQNBG-REOHCLBHSA-N L-serine Chemical compound OC[C@H](N)C(O)=O MTCFGRXMJLQNBG-REOHCLBHSA-N 0.000 description 5
- 108010003723 Single-Domain Antibodies Proteins 0.000 description 5
- DBMJMQXJHONAFJ-UHFFFAOYSA-M Sodium laurylsulphate Chemical compound [Na+].CCCCCCCCCCCCOS([O-])(=O)=O DBMJMQXJHONAFJ-UHFFFAOYSA-M 0.000 description 5
- 108010090804 Streptavidin Proteins 0.000 description 5
- 239000008346 aqueous phase Substances 0.000 description 5
- 238000006664 bond formation reaction Methods 0.000 description 5
- 230000000368 destabilizing Effects 0.000 description 5
- 210000004602 germ cell Anatomy 0.000 description 5
- 238000001802 infusion Methods 0.000 description 5
- 238000002347 injection Methods 0.000 description 5
- 239000007924 injection Substances 0.000 description 5
- 201000008968 osteosarcoma Diseases 0.000 description 5
- 230000003405 preventing Effects 0.000 description 5
- 238000002818 protein evolution Methods 0.000 description 5
- 238000005215 recombination Methods 0.000 description 5
- 238000011084 recovery Methods 0.000 description 5
- 238000000926 separation method Methods 0.000 description 5
- 238000006467 substitution reaction Methods 0.000 description 5
- 230000003612 virological Effects 0.000 description 5
- NFGXHKASABOEEW-UHFFFAOYSA-N (+)-methoprene Chemical compound COC(C)(C)CCCC(C)CC=CC(C)=CC(=O)OC(C)C NFGXHKASABOEEW-UHFFFAOYSA-N 0.000 description 4
- 206010000880 Acute myeloid leukaemia Diseases 0.000 description 4
- 241000271566 Aves Species 0.000 description 4
- 206010008958 Chronic lymphocytic leukaemia Diseases 0.000 description 4
- 108010047041 Complementarity Determining Regions Proteins 0.000 description 4
- 241000287828 Gallus gallus Species 0.000 description 4
- 210000004408 Hybridomas Anatomy 0.000 description 4
- 125000000510 L-tryptophano group Chemical group [H]C1=C([H])C([H])=C2N([H])C([H])=C(C([H])([H])[C@@]([H])(C(O[H])=O)N([H])[*])C2=C1[H] 0.000 description 4
- 208000000429 Leukemia, Lymphocytic, Chronic, B-Cell Diseases 0.000 description 4
- 208000008456 Leukemia, Myelogenous, Chronic, BCR-ABL Positive Diseases 0.000 description 4
- 208000007046 Leukemia, Myeloid, Acute Diseases 0.000 description 4
- 229910052765 Lutetium Inorganic materials 0.000 description 4
- 108010004729 Phycoerythrin Proteins 0.000 description 4
- 229920001213 Polysorbate 20 Polymers 0.000 description 4
- 229910052777 Praseodymium Inorganic materials 0.000 description 4
- 208000006664 Precursor Cell Lymphoblastic Leukemia-Lymphoma Diseases 0.000 description 4
- PYWVYCXTNDRMGF-UHFFFAOYSA-N Rhodamine B Chemical compound [Cl-].C=12C=CC(=[N+](CC)CC)C=C2OC2=CC(N(CC)CC)=CC=C2C=1C1=CC=CC=C1C(O)=O PYWVYCXTNDRMGF-UHFFFAOYSA-N 0.000 description 4
- 208000002474 Tinea Diseases 0.000 description 4
- ASCUXPQGEXGEMJ-GPLGTHOPSA-N [(2R,3S,4S,5R,6S)-3,4,5-triacetyloxy-6-[[(2R,3R,4S,5R,6R)-3,4,5-triacetyloxy-6-(4-methylanilino)oxan-2-yl]methoxy]oxan-2-yl]methyl acetate Chemical compound CC(=O)O[C@@H]1[C@@H](OC(C)=O)[C@@H](OC(C)=O)[C@@H](COC(=O)C)O[C@@H]1OC[C@@H]1[C@@H](OC(C)=O)[C@H](OC(C)=O)[C@@H](OC(C)=O)[C@H](NC=2C=CC(C)=CC=2)O1 ASCUXPQGEXGEMJ-GPLGTHOPSA-N 0.000 description 4
- 229910052767 actinium Inorganic materials 0.000 description 4
- 201000005510 acute lymphocytic leukemia Diseases 0.000 description 4
- 238000007792 addition Methods 0.000 description 4
- 125000003295 alanine group Chemical group N[C@@H](C)C(=O)* 0.000 description 4
- 230000004075 alteration Effects 0.000 description 4
- 229960000070 antineoplastic Monoclonal antibodies Drugs 0.000 description 4
- 229910052789 astatine Inorganic materials 0.000 description 4
- RYXHOMYVWAEKHL-UHFFFAOYSA-N astatine(.) Chemical compound [At] RYXHOMYVWAEKHL-UHFFFAOYSA-N 0.000 description 4
- 239000007795 chemical reaction product Substances 0.000 description 4
- 108091006028 chimera Proteins 0.000 description 4
- ZAMOUSCENKQFHK-UHFFFAOYSA-N chlorine atom Chemical compound [Cl] ZAMOUSCENKQFHK-UHFFFAOYSA-N 0.000 description 4
- 230000001684 chronic Effects 0.000 description 4
- 201000006934 chronic myeloid leukemia Diseases 0.000 description 4
- 230000021615 conjugation Effects 0.000 description 4
- 238000001816 cooling Methods 0.000 description 4
- 239000006185 dispersion Substances 0.000 description 4
- 238000005516 engineering process Methods 0.000 description 4
- 230000002349 favourable Effects 0.000 description 4
- 230000004927 fusion Effects 0.000 description 4
- 239000000499 gel Substances 0.000 description 4
- 230000003899 glycosylation Effects 0.000 description 4
- 238000006206 glycosylation reaction Methods 0.000 description 4
- 238000002744 homologous recombination Methods 0.000 description 4
- MHAJPDPJQMAIIY-UHFFFAOYSA-N hydrogen peroxide Chemical compound OO MHAJPDPJQMAIIY-UHFFFAOYSA-N 0.000 description 4
- APFVFJFRJDLVQX-UHFFFAOYSA-N indium Chemical compound [In] APFVFJFRJDLVQX-UHFFFAOYSA-N 0.000 description 4
- 229910052738 indium Inorganic materials 0.000 description 4
- 239000004615 ingredient Substances 0.000 description 4
- 238000007918 intramuscular administration Methods 0.000 description 4
- PNDPGZBMCMUPRI-UHFFFAOYSA-N iodine Chemical compound II PNDPGZBMCMUPRI-UHFFFAOYSA-N 0.000 description 4
- 229910052740 iodine Inorganic materials 0.000 description 4
- 239000011630 iodine Substances 0.000 description 4
- 239000011159 matrix material Substances 0.000 description 4
- 239000002609 media Substances 0.000 description 4
- 229960000060 monoclonal antibodies Drugs 0.000 description 4
- 238000007857 nested PCR Methods 0.000 description 4
- 239000002245 particle Substances 0.000 description 4
- 239000008194 pharmaceutical composition Substances 0.000 description 4
- 229910052698 phosphorus Inorganic materials 0.000 description 4
- 239000011574 phosphorus Substances 0.000 description 4
- 229920000642 polymer Polymers 0.000 description 4
- 235000010486 polyoxyethylene sorbitan monolaurate Nutrition 0.000 description 4
- PUDIUYLPXJFUGB-UHFFFAOYSA-N praseodymium Chemical compound [Pr] PUDIUYLPXJFUGB-UHFFFAOYSA-N 0.000 description 4
- 238000002360 preparation method Methods 0.000 description 4
- 238000000746 purification Methods 0.000 description 4
- 229910052702 rhenium Inorganic materials 0.000 description 4
- MHOVAHRLVXNVSD-UHFFFAOYSA-N rhodium Chemical compound [Rh] MHOVAHRLVXNVSD-UHFFFAOYSA-N 0.000 description 4
- 229910052703 rhodium Inorganic materials 0.000 description 4
- 239000010948 rhodium Substances 0.000 description 4
- BUGBHKTXTAQXES-UHFFFAOYSA-N selenium Chemical compound [Se] BUGBHKTXTAQXES-UHFFFAOYSA-N 0.000 description 4
- GKLVYJBZJHMRIY-UHFFFAOYSA-N technetium Chemical compound [Tc] GKLVYJBZJHMRIY-UHFFFAOYSA-N 0.000 description 4
- 229910052713 technetium Inorganic materials 0.000 description 4
- 229910052722 tritium Inorganic materials 0.000 description 4
- ORHBXUUXSCNDEV-UHFFFAOYSA-N umbelliferone Chemical compound C1=CC(=O)OC2=CC(O)=CC=C21 ORHBXUUXSCNDEV-UHFFFAOYSA-N 0.000 description 4
- 229920000160 (ribonucleotides)n+m Polymers 0.000 description 3
- 108010063905 Ampligase Proteins 0.000 description 3
- 244000303258 Annona diversifolia Species 0.000 description 3
- 235000002198 Annona diversifolia Nutrition 0.000 description 3
- 206010005098 Blastomycosis Diseases 0.000 description 3
- 206010007134 Candida infection Diseases 0.000 description 3
- 241000282693 Cercopithecidae Species 0.000 description 3
- 201000003884 Chagas disease Diseases 0.000 description 3
- 230000036499 Half live Effects 0.000 description 3
- 108010001336 Horseradish Peroxidase Proteins 0.000 description 3
- 206010021425 Immune system disease Diseases 0.000 description 3
- 208000007766 Kaposi Sarcoma Diseases 0.000 description 3
- 206010025323 Lymphomas Diseases 0.000 description 3
- 206010025650 Malignant melanoma Diseases 0.000 description 3
- 241001465754 Metazoa Species 0.000 description 3
- 241000699660 Mus musculus Species 0.000 description 3
- 206010025310 Other lymphomas Diseases 0.000 description 3
- 206010033128 Ovarian cancer Diseases 0.000 description 3
- 206010037660 Pyrexia Diseases 0.000 description 3
- 108020004511 Recombinant DNA Proteins 0.000 description 3
- 229920001914 Ribonucleotide Polymers 0.000 description 3
- 108010006785 Taq Polymerase Proteins 0.000 description 3
- 206010044269 Toxocariasis Diseases 0.000 description 3
- 241000893966 Trichophyton verrucosum Species 0.000 description 3
- 208000001756 Virus Disease Diseases 0.000 description 3
- 241000700605 Viruses Species 0.000 description 3
- 230000001154 acute Effects 0.000 description 3
- 230000000844 anti-bacterial Effects 0.000 description 3
- 230000001580 bacterial Effects 0.000 description 3
- JCXGWMGPZLAOME-UHFFFAOYSA-N bismuth Chemical compound [Bi] JCXGWMGPZLAOME-UHFFFAOYSA-N 0.000 description 3
- 229910052797 bismuth Inorganic materials 0.000 description 3
- 150000001768 cations Chemical class 0.000 description 3
- 239000000039 congener Substances 0.000 description 3
- 238000007796 conventional method Methods 0.000 description 3
- 239000005547 deoxyribonucleotide Substances 0.000 description 3
- 125000002637 deoxyribonucleotide group Chemical group 0.000 description 3
- 239000003937 drug carrier Substances 0.000 description 3
- 229940079593 drugs Drugs 0.000 description 3
- 238000004520 electroporation Methods 0.000 description 3
- 230000002255 enzymatic Effects 0.000 description 3
- 239000012530 fluid Substances 0.000 description 3
- 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 3
- 238000010438 heat treatment Methods 0.000 description 3
- 125000001165 hydrophobic group Chemical group 0.000 description 3
- 239000003446 ligand Substances 0.000 description 3
- 238000005259 measurement Methods 0.000 description 3
- 201000001441 melanoma Diseases 0.000 description 3
- 201000005962 mycosis fungoide Diseases 0.000 description 3
- 230000003287 optical Effects 0.000 description 3
- 239000000816 peptidomimetic Substances 0.000 description 3
- 238000006366 phosphorylation reaction Methods 0.000 description 3
- 230000000865 phosphorylative Effects 0.000 description 3
- 230000002265 prevention Effects 0.000 description 3
- 125000002924 primary amino group Chemical group [H]N([H])* 0.000 description 3
- 102000005962 receptors Human genes 0.000 description 3
- 108020003175 receptors Proteins 0.000 description 3
- 239000002336 ribonucleotide Substances 0.000 description 3
- 125000002652 ribonucleotide group Chemical group 0.000 description 3
- NINIDFKCEFEMDL-UHFFFAOYSA-N sulfur Chemical compound [S] NINIDFKCEFEMDL-UHFFFAOYSA-N 0.000 description 3
- 229910052727 yttrium Inorganic materials 0.000 description 3
- LLXVXPPXELIDGQ-UHFFFAOYSA-N (2,5-dioxopyrrolidin-1-yl) 3-(2,5-dioxopyrrol-1-yl)benzoate Chemical compound C=1C=CC(N2C(C=CC2=O)=O)=CC=1C(=O)ON1C(=O)CCC1=O LLXVXPPXELIDGQ-UHFFFAOYSA-N 0.000 description 2
- GUAHPAJOXVYFON-ZETCQYMHSA-N (8S)-8-azaniumyl-7-oxononanoate Chemical compound C[C@H](N)C(=O)CCCCCC(O)=O GUAHPAJOXVYFON-ZETCQYMHSA-N 0.000 description 2
- BACYUWVYYTXETD-UHFFFAOYSA-N 2-[dodecanoyl(methyl)amino]acetic acid Chemical compound CCCCCCCCCCCC(=O)N(C)CC(O)=O BACYUWVYYTXETD-UHFFFAOYSA-N 0.000 description 2
- HSTOKWSFWGCZMH-UHFFFAOYSA-N 3,3'-Diaminobenzidine Chemical compound C1=C(N)C(N)=CC=C1C1=CC=C(N)C(N)=C1 HSTOKWSFWGCZMH-UHFFFAOYSA-N 0.000 description 2
- 206010000565 Acquired immunodeficiency syndrome Diseases 0.000 description 2
- 241000251468 Actinopterygii Species 0.000 description 2
- 108010000239 Aequorin Proteins 0.000 description 2
- 210000000628 Antibody-Producing Cells Anatomy 0.000 description 2
- 208000003343 Antiphospholipid Syndrome Diseases 0.000 description 2
- 206010003571 Astrocytoma Diseases 0.000 description 2
- 101700072047 BCR Proteins 0.000 description 2
- 210000000988 Bone and Bones Anatomy 0.000 description 2
- 241000283690 Bos taurus Species 0.000 description 2
- 208000009899 Burkitt Lymphoma Diseases 0.000 description 2
- NLZUEZXRPGMBCV-UHFFFAOYSA-N Butylhydroxytoluene Chemical compound CC1=CC(C(C)(C)C)=C(O)C(C(C)(C)C)=C1 NLZUEZXRPGMBCV-UHFFFAOYSA-N 0.000 description 2
- 125000001433 C-terminal amino-acid group Chemical group 0.000 description 2
- 241000282832 Camelidae Species 0.000 description 2
- 206010007279 Carcinoid tumour of the gastrointestinal tract Diseases 0.000 description 2
- 208000010027 Carcinoma, Intraductal, Noninfiltrating Diseases 0.000 description 2
- 241000251730 Chondrichthyes Species 0.000 description 2
- 210000001106 Chromosomes, Artificial, Yeast Anatomy 0.000 description 2
- 229920000062 Coding strand Polymers 0.000 description 2
- 208000009798 Craniopharyngioma Diseases 0.000 description 2
- 208000003407 Creutzfeldt-Jakob Syndrome Diseases 0.000 description 2
- 201000010450 Creutzfeldt-Jakob disease Diseases 0.000 description 2
- 208000000307 Crimean Hemorrhagic Fever Diseases 0.000 description 2
- 201000003075 Crimean-Congo hemorrhagic fever Diseases 0.000 description 2
- 206010059547 Cutaneous larva migran Diseases 0.000 description 2
- IGXWBGJHJZYPQS-SSDOTTSWSA-N D-Luciferin Chemical compound OC(=O)[C@H]1CSC(C=2SC3=CC=C(O)C=C3N=2)=N1 IGXWBGJHJZYPQS-SSDOTTSWSA-N 0.000 description 2
- QIVBCDIJIAJPQS-SECBINFHSA-N D-tryptophane Chemical compound C1=CC=C2C(C[C@@H](N)C(O)=O)=CNC2=C1 QIVBCDIJIAJPQS-SECBINFHSA-N 0.000 description 2
- 102000016928 DNA-Directed DNA Polymerase Human genes 0.000 description 2
- 108010014303 DNA-Directed DNA Polymerase Proteins 0.000 description 2
- 108010015776 EC 1.1.3.4 Proteins 0.000 description 2
- 241000196324 Embryophyta Species 0.000 description 2
- 206010014881 Enterobiasis Diseases 0.000 description 2
- 208000006168 Ewing Sarcoma Diseases 0.000 description 2
- 229940116332 GLUCOSE OXIDASE Drugs 0.000 description 2
- GYHNNYVSQQEPJS-UHFFFAOYSA-N Gallium Chemical compound [Ga] GYHNNYVSQQEPJS-UHFFFAOYSA-N 0.000 description 2
- 206010072075 Gerstmann Straussler Scheinker syndrome Diseases 0.000 description 2
- 208000003736 Gerstmann-Straussler-Scheinker Disease Diseases 0.000 description 2
- 239000004366 Glucose oxidase Substances 0.000 description 2
- 206010018693 Granuloma inguinale Diseases 0.000 description 2
- 206010019143 Hantavirus pulmonary infection Diseases 0.000 description 2
- 208000007514 Herpes Zoster Diseases 0.000 description 2
- 241000238631 Hexapoda Species 0.000 description 2
- 241000282412 Homo Species 0.000 description 2
- 102000003839 Human Proteins Human genes 0.000 description 2
- 108090000144 Human Proteins Proteins 0.000 description 2
- 241000701806 Human papillomavirus Species 0.000 description 2
- 102000015434 Humanized Monoclonal Antibodies Human genes 0.000 description 2
- 108010064750 Humanized Monoclonal Antibodies Proteins 0.000 description 2
- 229960000310 ISOLEUCINE Drugs 0.000 description 2
- 206010027665 Immune disorder Diseases 0.000 description 2
- 206010073094 Intraductal proliferative breast lesion Diseases 0.000 description 2
- 206010061252 Intraocular melanoma Diseases 0.000 description 2
- FFEARJCKVFRZRR-BYPYZUCNSA-N L-methionine Chemical compound CSCC[C@H](N)C(O)=O FFEARJCKVFRZRR-BYPYZUCNSA-N 0.000 description 2
- KZSNJWFQEVHDMF-BYPYZUCNSA-N L-valine Chemical compound CC(C)[C@H](N)C(O)=O KZSNJWFQEVHDMF-BYPYZUCNSA-N 0.000 description 2
- 201000005099 Langerhans-cell histiocytosis Diseases 0.000 description 2
- 208000004023 Legionellosis Diseases 0.000 description 2
- 206010024324 Leukaemias Diseases 0.000 description 2
- 239000005089 Luciferase Substances 0.000 description 2
- 108060001084 Luciferase family Proteins 0.000 description 2
- HWYHZTIRURJOHG-UHFFFAOYSA-N Luminol Chemical compound O=C1NNC(=O)C2=C1C(N)=CC=C2 HWYHZTIRURJOHG-UHFFFAOYSA-N 0.000 description 2
- 206010025169 Lyme disease Diseases 0.000 description 2
- 208000003543 Lymphoma, T-Cell, Cutaneous Diseases 0.000 description 2
- 241000124008 Mammalia Species 0.000 description 2
- 208000008588 Molluscum Contagiosum Diseases 0.000 description 2
- ZTLGJPIZUOVDMT-UHFFFAOYSA-N N,N-dichlorotriazin-4-amine Chemical compound ClN(Cl)C1=CC=NN=N1 ZTLGJPIZUOVDMT-UHFFFAOYSA-N 0.000 description 2
- 208000002154 Non-Small-Cell Lung Carcinoma Diseases 0.000 description 2
- 108009000071 Non-small cell lung cancer Proteins 0.000 description 2
- 241000243985 Onchocerca volvulus Species 0.000 description 2
- 208000002042 Onchocerciasis Diseases 0.000 description 2
- 206010030861 Ophthalmia neonatorum Diseases 0.000 description 2
- 241000283898 Ovis Species 0.000 description 2
- 206010067517 Pancreatic neuroendocrine tumour Diseases 0.000 description 2
- 108010002747 Pfu DNA polymerase Proteins 0.000 description 2
- 208000005384 Pneumocystis Pneumonia Diseases 0.000 description 2
- 206010073755 Pneumocystis jirovecii pneumonia Diseases 0.000 description 2
- 241000288906 Primates Species 0.000 description 2
- 231100000654 Protein toxin Toxicity 0.000 description 2
- 208000000705 Rift Valley Fever Diseases 0.000 description 2
- 206010039207 Rocky mountain spotted fever Diseases 0.000 description 2
- 206010039491 Sarcoma Diseases 0.000 description 2
- 235000015076 Shorea robusta Nutrition 0.000 description 2
- 108010070144 Single-Chain Antibodies Proteins 0.000 description 2
- 102000005632 Single-Chain Antibodies Human genes 0.000 description 2
- KSAVQLQVUXSOCR-UHFFFAOYSA-M Sodium lauroyl sarcosinate Chemical compound [Na+].CCCCCCCCCCCC(=O)N(C)CC([O-])=O KSAVQLQVUXSOCR-UHFFFAOYSA-M 0.000 description 2
- 206010061372 Streptococcal infection Diseases 0.000 description 2
- 208000004441 Taeniasis Diseases 0.000 description 2
- 239000004473 Threonine Substances 0.000 description 2
- 208000009920 Trichuriasis Diseases 0.000 description 2
- 241000223109 Trypanosoma cruzi Species 0.000 description 2
- 206010061393 Typhus Diseases 0.000 description 2
- 210000002700 Urine Anatomy 0.000 description 2
- 208000001877 Whooping Cough Diseases 0.000 description 2
- 238000010521 absorption reaction Methods 0.000 description 2
- 150000007513 acids Chemical class 0.000 description 2
- 238000001042 affinity chromatography Methods 0.000 description 2
- 238000000246 agarose gel electrophoresis Methods 0.000 description 2
- 230000003281 allosteric Effects 0.000 description 2
- 230000000845 anti-microbial Effects 0.000 description 2
- 230000000890 antigenic Effects 0.000 description 2
- 125000004429 atoms Chemical group 0.000 description 2
- 201000008271 atypical teratoid rhabdoid tumor Diseases 0.000 description 2
- 230000003115 biocidal Effects 0.000 description 2
- 230000001413 cellular Effects 0.000 description 2
- 239000002738 chelating agent Substances 0.000 description 2
- VEXZGXHMUGYJMC-UHFFFAOYSA-M chloride anion Chemical compound [Cl-] VEXZGXHMUGYJMC-UHFFFAOYSA-M 0.000 description 2
- 239000000460 chlorine Substances 0.000 description 2
- 229910052801 chlorine Inorganic materials 0.000 description 2
- VYZAMTAEIAYCRO-UHFFFAOYSA-N chromium Chemical compound [Cr] VYZAMTAEIAYCRO-UHFFFAOYSA-N 0.000 description 2
- 229910052804 chromium Inorganic materials 0.000 description 2
- 239000011651 chromium Substances 0.000 description 2
- 238000007374 clinical diagnostic method Methods 0.000 description 2
- GUTLYIVDDKVIGB-UHFFFAOYSA-N cobalt Chemical compound [Co] GUTLYIVDDKVIGB-UHFFFAOYSA-N 0.000 description 2
- 229910052803 cobalt Inorganic materials 0.000 description 2
- 239000010941 cobalt Substances 0.000 description 2
- 201000009230 common cold Diseases 0.000 description 2
- 230000000875 corresponding Effects 0.000 description 2
- 230000001808 coupling Effects 0.000 description 2
- 238000010168 coupling process Methods 0.000 description 2
- 238000005859 coupling reaction Methods 0.000 description 2
- 238000004132 cross linking Methods 0.000 description 2
- 201000007241 cutaneous T cell lymphoma Diseases 0.000 description 2
- 125000000151 cysteine group Chemical group N[C@@H](CS)C(=O)* 0.000 description 2
- 230000001085 cytostatic Effects 0.000 description 2
- 239000000824 cytostatic agent Substances 0.000 description 2
- 230000001472 cytotoxic Effects 0.000 description 2
- 231100000433 cytotoxic Toxicity 0.000 description 2
- 238000001212 derivatisation Methods 0.000 description 2
- 238000010586 diagram Methods 0.000 description 2
- 239000002612 dispersion media Substances 0.000 description 2
- 239000010459 dolomite Substances 0.000 description 2
- 229910000514 dolomite Inorganic materials 0.000 description 2
- 201000007273 ductal carcinoma in situ Diseases 0.000 description 2
- 238000010828 elution Methods 0.000 description 2
- 210000003527 eukaryotic cell Anatomy 0.000 description 2
- 201000006353 filariasis Diseases 0.000 description 2
- 238000001943 fluorescence-activated cell sorting Methods 0.000 description 2
- 230000005714 functional activity Effects 0.000 description 2
- 230000002538 fungal Effects 0.000 description 2
- 201000011243 gastrointestinal stromal tumor Diseases 0.000 description 2
- 230000002068 genetic Effects 0.000 description 2
- 235000019420 glucose oxidase Nutrition 0.000 description 2
- 201000009277 hairy cell leukemia Diseases 0.000 description 2
- 201000006215 hand, foot and mouth disease Diseases 0.000 description 2
- 201000005648 hantavirus pulmonary syndrome Diseases 0.000 description 2
- 201000001066 hemolytic-uremic syndrome Diseases 0.000 description 2
- 201000000907 hemorrhagic fever with renal syndrome Diseases 0.000 description 2
- 201000009163 human granulocytic anaplasmosis Diseases 0.000 description 2
- 230000002519 immonomodulatory Effects 0.000 description 2
- 238000001114 immunoprecipitation Methods 0.000 description 2
- 238000007912 intraperitoneal administration Methods 0.000 description 2
- 229910052742 iron Inorganic materials 0.000 description 2
- 201000002529 islet cell tumor Diseases 0.000 description 2
- 230000000670 limiting Effects 0.000 description 2
- 201000006721 lip cancer Diseases 0.000 description 2
- 239000002502 liposome Substances 0.000 description 2
- 239000006166 lysate Substances 0.000 description 2
- 239000003550 marker Substances 0.000 description 2
- 230000001404 mediated Effects 0.000 description 2
- 229910052751 metal Inorganic materials 0.000 description 2
- 239000002184 metal Substances 0.000 description 2
- 200000000016 middle east respiratory syndrome Diseases 0.000 description 2
- 230000003278 mimic Effects 0.000 description 2
- 201000007224 myeloproliferative neoplasm Diseases 0.000 description 2
- 229920002113 octoxynol Polymers 0.000 description 2
- 201000002575 ocular melanoma Diseases 0.000 description 2
- 201000005443 oral cavity cancer Diseases 0.000 description 2
- 210000000056 organs Anatomy 0.000 description 2
- 201000005702 pertussis Diseases 0.000 description 2
- 239000008177 pharmaceutical agent Substances 0.000 description 2
- 201000000317 pneumocystosis Diseases 0.000 description 2
- 229920002704 polyhistidine Polymers 0.000 description 2
- 238000003752 polymerase chain reaction Methods 0.000 description 2
- 238000006116 polymerization reaction Methods 0.000 description 2
- 239000000843 powder Substances 0.000 description 2
- 210000001938 protoplasts Anatomy 0.000 description 2
- 230000000241 respiratory Effects 0.000 description 2
- 201000000582 retinoblastoma Diseases 0.000 description 2
- 238000003757 reverse transcription PCR Methods 0.000 description 2
- 201000009410 rhabdomyosarcoma Diseases 0.000 description 2
- 229940016590 sarkosyl Drugs 0.000 description 2
- 229910052711 selenium Inorganic materials 0.000 description 2
- 239000011669 selenium Substances 0.000 description 2
- 201000005113 shigellosis Diseases 0.000 description 2
- 201000000849 skin cancer Diseases 0.000 description 2
- FAPWRFPIFSIZLT-UHFFFAOYSA-M sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 2
- 239000011780 sodium chloride Substances 0.000 description 2
- 229940045885 sodium lauroyl sarcosinate Drugs 0.000 description 2
- 239000002904 solvent Substances 0.000 description 2
- 230000000392 somatic Effects 0.000 description 2
- 229910052717 sulfur Inorganic materials 0.000 description 2
- 239000011593 sulfur Substances 0.000 description 2
- 239000006228 supernatant Substances 0.000 description 2
- 239000000725 suspension Substances 0.000 description 2
- 239000003826 tablet Substances 0.000 description 2
- 201000010618 tinea cruris Diseases 0.000 description 2
- 201000004647 tinea pedis Diseases 0.000 description 2
- 210000001519 tissues Anatomy 0.000 description 2
- 238000010361 transduction Methods 0.000 description 2
- 230000026683 transduction Effects 0.000 description 2
- 238000001890 transfection Methods 0.000 description 2
- 101710039969 tth1 Proteins 0.000 description 2
- 239000004474 valine Substances 0.000 description 2
- 239000003981 vehicle Substances 0.000 description 2
- 201000000752 white piedra Diseases 0.000 description 2
- XPDXVDYUQZHFPV-UHFFFAOYSA-N 5-Dimethylaminonaphthyl-5-sulfonyl chloride Chemical compound C1=CC=C2C(N(C)C)=CC=CC2=C1S(Cl)(=O)=O XPDXVDYUQZHFPV-UHFFFAOYSA-N 0.000 description 1
- 208000002008 AIDS-Related Lymphoma Diseases 0.000 description 1
- 101710034857 ATIC Proteins 0.000 description 1
- 206010063409 Acarodermatitis Diseases 0.000 description 1
- 229940022698 Acetylcholinesterase Drugs 0.000 description 1
- 102000012440 Acetylcholinesterase Human genes 0.000 description 1
- 108010022752 Acetylcholinesterase Proteins 0.000 description 1
- 206010051894 Acinetobacter infection Diseases 0.000 description 1
- 201000004304 Addison's disease Diseases 0.000 description 1
- 208000007128 Adrenocortical Carcinoma Diseases 0.000 description 1
- 206010001433 Aesthesioneuroblastoma Diseases 0.000 description 1
- 208000000230 African Trypanosomiasis Diseases 0.000 description 1
- 208000008190 Agammaglobulinemia Diseases 0.000 description 1
- 102000002260 Alkaline Phosphatase Human genes 0.000 description 1
- 108020004774 Alkaline Phosphatase Proteins 0.000 description 1
- 208000004631 Alopecia Areata Diseases 0.000 description 1
- 208000004881 Amebiasis Diseases 0.000 description 1
- 206010001935 American trypanosomiasis Diseases 0.000 description 1
- 206010001980 Amoebiasis Diseases 0.000 description 1
- 206010002023 Amyloidosis Diseases 0.000 description 1
- 206010002022 Amyloidosis Diseases 0.000 description 1
- 206010061424 Anal cancer Diseases 0.000 description 1
- 208000006730 Anaplasmosis Diseases 0.000 description 1
- 208000005067 Anisakiasis Diseases 0.000 description 1
- 229940064005 Antibiotic throat preparations Drugs 0.000 description 1
- 229940083879 Antibiotics FOR TREATMENT OF HEMORRHOIDS AND ANAL FISSURES FOR TOPICAL USE Drugs 0.000 description 1
- 229940042052 Antibiotics for systemic use Drugs 0.000 description 1
- 229940042786 Antitubercular Antibiotics Drugs 0.000 description 1
- 206010073360 Appendix cancer Diseases 0.000 description 1
- 241001135700 Arcanobacterium haemolyticum Species 0.000 description 1
- 108020004634 Archaeal DNA Proteins 0.000 description 1
- 201000009695 Argentine hemorrhagic fever Diseases 0.000 description 1
- 239000004475 Arginine Substances 0.000 description 1
- 208000008715 Ascaridida Infections Diseases 0.000 description 1
- 229960001230 Asparagine Drugs 0.000 description 1
- 229960005261 Aspartic Acid Drugs 0.000 description 1
- 206010003486 Aspergillus infection Diseases 0.000 description 1
- 206010003827 Autoimmune hepatitis Diseases 0.000 description 1
- 108091008154 B cell receptors Proteins 0.000 description 1
- 206010055181 BK virus infection Diseases 0.000 description 1
- 208000004429 Bacillary Dysentery Diseases 0.000 description 1
- 241000193738 Bacillus anthracis Species 0.000 description 1
- 229940075612 Bacillus cereus Drugs 0.000 description 1
- 241000193755 Bacillus cereus Species 0.000 description 1
- 208000004926 Bacterial Vaginosis Diseases 0.000 description 1
- 206010060945 Bacterial infection Diseases 0.000 description 1
- 206010058890 Bacteroides infection Diseases 0.000 description 1
- 208000007456 Balantidiasis Diseases 0.000 description 1
- 206010004145 Bartonellosis Diseases 0.000 description 1
- 206010004146 Basal cell carcinoma Diseases 0.000 description 1
- 208000001119 Benign Fibrous Histiocytoma Diseases 0.000 description 1
- 206010004593 Bile duct cancer Diseases 0.000 description 1
- 108010071919 Bispecific Antibodies Proteins 0.000 description 1
- 206010005003 Bladder cancer Diseases 0.000 description 1
- 206010005913 Body tinea Diseases 0.000 description 1
- 201000009694 Bolivian hemorrhagic fever Diseases 0.000 description 1
- 206010005949 Bone cancer Diseases 0.000 description 1
- 208000003508 Botulism Diseases 0.000 description 1
- 241000701822 Bovine papillomavirus Species 0.000 description 1
- 208000003174 Brain Neoplasms Diseases 0.000 description 1
- 206010006143 Brain stem glioma Diseases 0.000 description 1
- 201000010424 Brazilian hemorrhagic fever Diseases 0.000 description 1
- 206010006187 Breast cancer Diseases 0.000 description 1
- 206010006500 Brucellosis Diseases 0.000 description 1
- 208000000594 Bullous Pemphigoid Diseases 0.000 description 1
- 206010073031 Burkholderia infection Diseases 0.000 description 1
- 206010069748 Burkholderia pseudomallei infection Diseases 0.000 description 1
- 208000006448 Buruli Ulcer Diseases 0.000 description 1
- 108060001070 CACNA1F Proteins 0.000 description 1
- 102100017588 CACNA1F Human genes 0.000 description 1
- 101700018538 CDR4 Proteins 0.000 description 1
- 210000003229 CMP Anatomy 0.000 description 1
- 102100005552 CR1 Human genes 0.000 description 1
- 101700042195 CR1 Proteins 0.000 description 1
- 241000222122 Candida albicans Species 0.000 description 1
- 206010007187 Capillariasis Diseases 0.000 description 1
- 102000004040 Capsid Proteins Human genes 0.000 description 1
- 108090000565 Capsid Proteins Proteins 0.000 description 1
- 208000002458 Carcinoid Tumor Diseases 0.000 description 1
- 206010007275 Carcinoid tumour Diseases 0.000 description 1
- 208000008787 Cardiovascular Disease Diseases 0.000 description 1
- 201000011310 Carrion's disease Diseases 0.000 description 1
- 208000003732 Cat-Scratch Disease Diseases 0.000 description 1
- 241000700198 Cavia Species 0.000 description 1
- 210000000170 Cell Membrane Anatomy 0.000 description 1
- 206010007882 Cellulitis Diseases 0.000 description 1
- 241000606161 Chlamydia Species 0.000 description 1
- 241001647372 Chlamydia pneumoniae Species 0.000 description 1
- 206010008631 Cholera Diseases 0.000 description 1
- 102000003914 Cholinesterases Human genes 0.000 description 1
- 108090000322 Cholinesterases Proteins 0.000 description 1
- 206010008761 Choriomeningitis lymphocytic Diseases 0.000 description 1
- 210000000349 Chromosomes Anatomy 0.000 description 1
- 230000037250 Clearance Effects 0.000 description 1
- 206010009344 Clonorchiasis Diseases 0.000 description 1
- 241001112695 Clostridiales Species 0.000 description 1
- 206010009657 Clostridium difficile colitis Diseases 0.000 description 1
- 229940038704 Clostridium perfringens Drugs 0.000 description 1
- 241000223205 Coccidioides immitis Species 0.000 description 1
- 206010009839 Coeliac disease Diseases 0.000 description 1
- 102000008186 Collagen Human genes 0.000 description 1
- 108010035532 Collagen Proteins 0.000 description 1
- 206010009944 Colon cancer Diseases 0.000 description 1
- 239000004971 Cross linker Substances 0.000 description 1
- 208000008953 Cryptosporidiosis Diseases 0.000 description 1
- 206010011502 Cryptosporidiosis infection Diseases 0.000 description 1
- 206010061802 Cyclosporidium infection Diseases 0.000 description 1
- 206010011831 Cytomegalovirus infection Diseases 0.000 description 1
- 208000001490 Dengue Diseases 0.000 description 1
- 206010012310 Dengue fever Diseases 0.000 description 1
- 241000305506 Desmodesmus Species 0.000 description 1
- 206010012601 Diabetes mellitus Diseases 0.000 description 1
- 206010013023 Diphtheria Diseases 0.000 description 1
- 206010013029 Diphyllobothriasis Diseases 0.000 description 1
- ZWIBGKZDAWNIFC-UHFFFAOYSA-N Disuccinimidyl suberate Chemical compound O=C1CCC(=O)N1OC(=O)CCCCCCC(=O)ON1C(=O)CCC1=O ZWIBGKZDAWNIFC-UHFFFAOYSA-N 0.000 description 1
- 208000008576 Dracunculiasis Diseases 0.000 description 1
- 241000710945 Eastern equine encephalitis virus Species 0.000 description 1
- 206010014071 Ebola disease Diseases 0.000 description 1
- 201000011001 Ebola hemorrhagic fever Diseases 0.000 description 1
- 206010014096 Echinococciasis Diseases 0.000 description 1
- 208000009366 Echinococcosis Diseases 0.000 description 1
- 208000000292 Ehrlichiosis Diseases 0.000 description 1
- 208000006036 Elephantiasis Diseases 0.000 description 1
- 206010014599 Encephalitis Diseases 0.000 description 1
- 206010014611 Encephalitis venezuelan equine Diseases 0.000 description 1
- 206010014733 Endometrial cancer Diseases 0.000 description 1
- 208000004232 Enteritis Diseases 0.000 description 1
- 241000194033 Enterococcus Species 0.000 description 1
- 206010014909 Enterovirus infection Diseases 0.000 description 1
- 206010014967 Ependymoma Diseases 0.000 description 1
- 206010014979 Epidemic typhus Diseases 0.000 description 1
- 208000007985 Erythema Infectiosum Diseases 0.000 description 1
- 241000701959 Escherichia virus Lambda Species 0.000 description 1
- 229910052693 Europium Inorganic materials 0.000 description 1
- 208000010201 Exanthema Diseases 0.000 description 1
- 206010016235 Fasciolopsiasis Diseases 0.000 description 1
- 206010053717 Fibrous histiocytoma Diseases 0.000 description 1
- 241000710831 Flavivirus Species 0.000 description 1
- 206010016952 Food poisoning Diseases 0.000 description 1
- 210000002683 Foot Anatomy 0.000 description 1
- ZZUFCTLCJUWOSV-UHFFFAOYSA-N Furosemide Chemical compound C1=C(Cl)C(S(=O)(=O)N)=CC(C(O)=O)=C1NCC1=CC=CO1 ZZUFCTLCJUWOSV-UHFFFAOYSA-N 0.000 description 1
- 206010017564 Fusobacterium infection Diseases 0.000 description 1
- 206010017915 Gastroenteritis shigella Diseases 0.000 description 1
- 206010017916 Gastroenteritis staphylococcal Diseases 0.000 description 1
- 108010010803 Gelatin Proteins 0.000 description 1
- 210000004907 Glands Anatomy 0.000 description 1
- 206010018338 Glioma Diseases 0.000 description 1
- 229960002989 Glutamic Acid Drugs 0.000 description 1
- 208000000807 Gnathostomiasis Diseases 0.000 description 1
- 208000001786 Gonorrhea Diseases 0.000 description 1
- 206010018612 Gonorrhoea Diseases 0.000 description 1
- 229940093922 Gynecological Antibiotics Drugs 0.000 description 1
- 206010019375 Helicobacter infection Diseases 0.000 description 1
- 208000006750 Hematuria Diseases 0.000 description 1
- 208000005252 Hepatitis A Diseases 0.000 description 1
- 208000002672 Hepatitis B Diseases 0.000 description 1
- 208000005176 Hepatitis C Diseases 0.000 description 1
- 208000005331 Hepatitis D Diseases 0.000 description 1
- 208000009889 Herpes Simplex Diseases 0.000 description 1
- 206010020243 Hodgkin's disease Diseases 0.000 description 1
- 201000006743 Hodgkin's lymphoma Diseases 0.000 description 1
- 206010020376 Hookworm infection Diseases 0.000 description 1
- 241000046923 Human bocavirus Species 0.000 description 1
- 241000701044 Human gammaherpesvirus 4 Species 0.000 description 1
- 241000342334 Human metapneumovirus Species 0.000 description 1
- 208000007188 Hymenolepiasis Diseases 0.000 description 1
- 206010020751 Hypersensitivity Diseases 0.000 description 1
- 206010020772 Hypertension Diseases 0.000 description 1
- 206010020983 Hypogammaglobulinaemia Diseases 0.000 description 1
- 229940072221 IMMUNOGLOBULINS Drugs 0.000 description 1
- 108010054477 Immunoglobulin Fab Fragments Proteins 0.000 description 1
- 102000001706 Immunoglobulin Fab Fragments Human genes 0.000 description 1
- 102000012745 Immunoglobulin Subunits Human genes 0.000 description 1
- 108010079585 Immunoglobulin Subunits Proteins 0.000 description 1
- 206010022000 Influenza Diseases 0.000 description 1
- 206010061524 Inner ear disease Diseases 0.000 description 1
- 206010022437 Insomnia Diseases 0.000 description 1
- 229920002459 Intron Polymers 0.000 description 1
- 108020004391 Introns Proteins 0.000 description 1
- 241000726306 Irus Species 0.000 description 1
- 206010023076 Isosporiasis Diseases 0.000 description 1
- 241000229754 Iva xanthiifolia Species 0.000 description 1
- 201000007313 Kawasaki disease Diseases 0.000 description 1
- 206010023332 Keratitis Diseases 0.000 description 1
- 241000589014 Kingella kingae Species 0.000 description 1
- 206010023497 Kuru Diseases 0.000 description 1
- QNAYBMKLOCPYGJ-REOHCLBHSA-N L-alanine Chemical compound C[C@H](N)C(O)=O QNAYBMKLOCPYGJ-REOHCLBHSA-N 0.000 description 1
- DCXYFEDJOCDNAF-REOHCLBHSA-N L-asparagine Chemical compound OC(=O)[C@@H](N)CC(N)=O DCXYFEDJOCDNAF-REOHCLBHSA-N 0.000 description 1
- CKLJMWTZIZZHCS-REOHCLBHSA-N L-aspartic acid Chemical compound OC(=O)[C@@H](N)CC(O)=O CKLJMWTZIZZHCS-REOHCLBHSA-N 0.000 description 1
- WHUUTDBJXJRKMK-VKHMYHEASA-N L-glutamic acid Chemical compound OC(=O)[C@@H](N)CCC(O)=O WHUUTDBJXJRKMK-VKHMYHEASA-N 0.000 description 1
- ZDXPYRJPNDTMRX-VKHMYHEASA-N L-glutamine Chemical compound OC(=O)[C@@H](N)CCC(N)=O ZDXPYRJPNDTMRX-VKHMYHEASA-N 0.000 description 1
- HNDVDQJCIGZPNO-YFKPBYRVSA-N L-histidine Chemical compound OC(=O)[C@@H](N)CC1=CN=CN1 HNDVDQJCIGZPNO-YFKPBYRVSA-N 0.000 description 1
- ONIBWKKTOPOVIA-BYPYZUCNSA-N L-proline Chemical compound OC(=O)[C@@H]1CCCN1 ONIBWKKTOPOVIA-BYPYZUCNSA-N 0.000 description 1
- 208000004501 Labyrinth Disease Diseases 0.000 description 1
- 241000282852 Lama guanicoe Species 0.000 description 1
- 208000004204 Larva Migrans Diseases 0.000 description 1
- 206010023825 Laryngeal cancer Diseases 0.000 description 1
- 206010023927 Lassa fever Diseases 0.000 description 1
- 208000007764 Legionnaires' Disease Diseases 0.000 description 1
- 208000004554 Leishmaniasis Diseases 0.000 description 1
- 206010024229 Leprosy Diseases 0.000 description 1
- 206010024238 Leptospirosis Diseases 0.000 description 1
- 206010061523 Lip and/or oral cavity cancer Diseases 0.000 description 1
- 210000004185 Liver Anatomy 0.000 description 1
- 206010058467 Lung neoplasm malignant Diseases 0.000 description 1
- 208000001419 Lymphocytic Choriomeningitis Diseases 0.000 description 1
- 206010025312 Lymphoma AIDS related Diseases 0.000 description 1
- 239000004472 Lysine Substances 0.000 description 1
- 206010025482 Malaise Diseases 0.000 description 1
- 208000006644 Malignant Fibrous Histiocytoma Diseases 0.000 description 1
- 206010025557 Malignant fibrous histiocytoma of bone Diseases 0.000 description 1
- 208000002030 Merkel Cell Carcinoma Diseases 0.000 description 1
- 206010027406 Mesothelioma Diseases 0.000 description 1
- 208000008466 Metabolic Disease Diseases 0.000 description 1
- 206010066226 Metapneumovirus infection Diseases 0.000 description 1
- 206010061289 Metastatic neoplasm Diseases 0.000 description 1
- 241000840267 Moma Species 0.000 description 1
- 208000005871 Monkeypox Diseases 0.000 description 1
- 241000713333 Mouse mammary tumor virus Species 0.000 description 1
- 208000001725 Mucocutaneous Lymph Node Syndrome Diseases 0.000 description 1
- 206010028193 Multiple endocrine neoplasia syndrome Diseases 0.000 description 1
- 208000005647 Mumps Diseases 0.000 description 1
- 206010028282 Murine typhus Diseases 0.000 description 1
- 241000699670 Mus sp. Species 0.000 description 1
- 241000238367 Mya arenaria Species 0.000 description 1
- 206010066289 Mycobacterium ulcerans infection Diseases 0.000 description 1
- 208000001572 Mycoplasma Pneumonia Diseases 0.000 description 1
- 201000008235 Mycoplasma pneumoniae pneumonia Diseases 0.000 description 1
- 208000006123 Myiasis Diseases 0.000 description 1
- 210000003928 Nasal Cavity Anatomy 0.000 description 1
- 206010028729 Nasal cavity cancer Diseases 0.000 description 1
- 206010028767 Nasal sinus cancer Diseases 0.000 description 1
- 206010061306 Nasopharyngeal cancer Diseases 0.000 description 1
- 206010029260 Neuroblastoma Diseases 0.000 description 1
- 206010029266 Neuroendocrine carcinoma of the skin Diseases 0.000 description 1
- BAWFJGJZGIEFAR-NNYOXOHSSA-N Nicotinamide adenine dinucleotide Chemical compound NC(=O)C1=CC=C[N+]([C@H]2[C@@H]([C@H](O)[C@@H](COP([O-])(=O)OP(O)(=O)OC[C@@H]3[C@H]([C@@H](O)[C@@H](O3)N3C4=NC=NC(N)=C4N=C3)O)O2)O)=C1 BAWFJGJZGIEFAR-NNYOXOHSSA-N 0.000 description 1
- 206010029444 Nocardiosis Diseases 0.000 description 1
- 206010029592 Non-Hodgkin's lymphomas Diseases 0.000 description 1
- 241001263478 Norovirus Species 0.000 description 1
- 108091005503 Nucleic proteins Chemical group 0.000 description 1
- 208000003177 Ocular Onchocerciasis Diseases 0.000 description 1
- 206010030113 Oedema Diseases 0.000 description 1
- 206010030155 Oesophageal carcinoma Diseases 0.000 description 1
- 208000000160 Olfactory Esthesioneuroblastoma Diseases 0.000 description 1
- 208000010195 Onychomycosis Diseases 0.000 description 1
- 208000003692 Opisthorchiasis Diseases 0.000 description 1
- 208000007027 Oral Candidiasis Diseases 0.000 description 1
- 206010031096 Oropharyngeal cancer Diseases 0.000 description 1
- 208000002193 Pain Diseases 0.000 description 1
- 208000008443 Pancreatic Carcinoma Diseases 0.000 description 1
- 208000003154 Papilloma Diseases 0.000 description 1
- 206010033767 Paracoccidioides infection Diseases 0.000 description 1
- 208000007312 Paraganglioma Diseases 0.000 description 1
- 206010061332 Paraganglion neoplasm Diseases 0.000 description 1
- 206010033794 Paragonimiasis Diseases 0.000 description 1
- 210000003695 Paranasal Sinuses Anatomy 0.000 description 1
- 241000517324 Pediculidae Species 0.000 description 1
- 241000517307 Pediculus humanus Species 0.000 description 1
- 201000000376 Pediculus humanus capitis infestation Diseases 0.000 description 1
- 206010034277 Pemphigoid Diseases 0.000 description 1
- 206010034299 Penile cancer Diseases 0.000 description 1
- 229940072417 Peroxidase Drugs 0.000 description 1
- 108090000437 Peroxidases Proteins 0.000 description 1
- 102000003992 Peroxidases Human genes 0.000 description 1
- 206010034800 Phaeochromocytoma Diseases 0.000 description 1
- 206010034811 Pharyngeal cancer Diseases 0.000 description 1
- 229960005190 Phenylalanine Drugs 0.000 description 1
- 241001674048 Phthiraptera Species 0.000 description 1
- 208000007913 Pituitary Neoplasms Diseases 0.000 description 1
- 241000101040 Pityriasis Species 0.000 description 1
- 206010035148 Plague Diseases 0.000 description 1
- 210000004011 Plasma Cells Anatomy 0.000 description 1
- 206010035226 Plasma cell myeloma Diseases 0.000 description 1
- 206010035664 Pneumonia Diseases 0.000 description 1
- 206010035737 Pneumonia viral Diseases 0.000 description 1
- 229920002732 Polyanhydride Polymers 0.000 description 1
- 229920000954 Polyglycolide Polymers 0.000 description 1
- 241001505332 Polyomavirus sp. Species 0.000 description 1
- 206010054161 Pontiac fever Diseases 0.000 description 1
- 241000605861 Prevotella Species 0.000 description 1
- 206010036807 Progressive multifocal leukoencephalopathy Diseases 0.000 description 1
- 206010060862 Prostate cancer Diseases 0.000 description 1
- 208000003251 Pruritus Diseases 0.000 description 1
- 206010037151 Psittacosis Diseases 0.000 description 1
- 241000517305 Pthiridae Species 0.000 description 1
- 201000004360 Pthirus pubis infestation Diseases 0.000 description 1
- 206010037688 Q fever Diseases 0.000 description 1
- 206010037742 Rabies Diseases 0.000 description 1
- 206010037844 Rash Diseases 0.000 description 1
- 108060006943 RdRp Proteins 0.000 description 1
- 206010038038 Rectal cancer Diseases 0.000 description 1
- 208000007865 Relapsing Fever Diseases 0.000 description 1
- 208000006265 Renal Cell Carcinoma Diseases 0.000 description 1
- 206010038389 Renal cancer Diseases 0.000 description 1
- 206010061603 Respiratory syncytial virus infection Diseases 0.000 description 1
- 208000004364 Rhinosporidiosis Diseases 0.000 description 1
- 206010061494 Rhinovirus infection Diseases 0.000 description 1
- 241000606723 Rickettsia akari Species 0.000 description 1
- 206010067470 Rotavirus infection Diseases 0.000 description 1
- 241000714474 Rous sarcoma virus Species 0.000 description 1
- 241000315672 SARS coronavirus Species 0.000 description 1
- 206010039447 Salmonellosis Diseases 0.000 description 1
- 241000369757 Sapovirus Species 0.000 description 1
- 208000005687 Scabies Diseases 0.000 description 1
- 241000447727 Scabies Species 0.000 description 1
- 210000004761 Scalp Anatomy 0.000 description 1
- 241000710961 Semliki Forest virus Species 0.000 description 1
- 210000002966 Serum Anatomy 0.000 description 1
- 206010040550 Shigella infection Diseases 0.000 description 1
- 208000000587 Small Cell Lung Carcinoma Diseases 0.000 description 1
- 206010041067 Small cell lung cancer Diseases 0.000 description 1
- 208000001203 Smallpox Diseases 0.000 description 1
- 206010041736 Sporotrichosis Diseases 0.000 description 1
- 206010041823 Squamous cell carcinoma Diseases 0.000 description 1
- 208000008582 Staphylococcal Food Poisoning Diseases 0.000 description 1
- 206010041925 Staphylococcal infection Diseases 0.000 description 1
- 206010042254 Strongyloidiasis Diseases 0.000 description 1
- 229940035718 Sular Drugs 0.000 description 1
- 231100000617 Superantigen Toxicity 0.000 description 1
- 241000282890 Sus Species 0.000 description 1
- 208000006379 Syphilis Diseases 0.000 description 1
- 210000003283 T-Lymphocytes, Helper-Inducer Anatomy 0.000 description 1
- 206010057644 Testis cancer Diseases 0.000 description 1
- 206010043376 Tetanus Diseases 0.000 description 1
- 206010043515 Throat cancer Diseases 0.000 description 1
- 208000008732 Thymoma Diseases 0.000 description 1
- 241000130764 Tinea Species 0.000 description 1
- 208000007712 Tinea Versicolor Diseases 0.000 description 1
- 206010043865 Tinea blanca Diseases 0.000 description 1
- 206010043866 Tinea capitis Diseases 0.000 description 1
- 206010043871 Tinea nigra Diseases 0.000 description 1
- 206010056131 Tinea versicolour Diseases 0.000 description 1
- 229940024982 Topical Antifungal Antibiotics Drugs 0.000 description 1
- 206010044325 Trachoma Diseases 0.000 description 1
- 241000390203 Trachoma Species 0.000 description 1
- 206010044407 Transitional cell cancer of the renal pelvis and ureter Diseases 0.000 description 1
- 206010052779 Transplant rejections Diseases 0.000 description 1
- QORWJWZARLRLPR-UHFFFAOYSA-H Tricalcium phosphate Chemical compound [Ca+2].[Ca+2].[Ca+2].[O-]P([O-])([O-])=O.[O-]P([O-])([O-])=O QORWJWZARLRLPR-UHFFFAOYSA-H 0.000 description 1
- 208000003982 Trichinellosis Diseases 0.000 description 1
- 206010044608 Trichiniasis Diseases 0.000 description 1
- 208000005448 Trichomonas Infections Diseases 0.000 description 1
- 206010044620 Trichomoniasis Diseases 0.000 description 1
- HRXKRNGNAMMEHJ-UHFFFAOYSA-K Trisodium citrate Chemical compound [Na+].[Na+].[Na+].[O-]C(=O)CC(O)(CC([O-])=O)C([O-])=O HRXKRNGNAMMEHJ-UHFFFAOYSA-K 0.000 description 1
- 241000223105 Trypanosoma brucei Species 0.000 description 1
- 206010045146 Tularaemia Diseases 0.000 description 1
- 241000287411 Turdidae Species 0.000 description 1
- 241000202921 Ureaplasma urealyticum Species 0.000 description 1
- 206010046431 Urethral cancer Diseases 0.000 description 1
- 206010046766 Uterine cancer Diseases 0.000 description 1
- 241000700618 Vaccinia virus Species 0.000 description 1
- 206010046885 Vaginal cancer Diseases 0.000 description 1
- 206010062167 Vaginitis bacterial Diseases 0.000 description 1
- 206010046980 Varicella Diseases 0.000 description 1
- 241000870995 Variola Species 0.000 description 1
- 241000700647 Variola virus Species 0.000 description 1
- 208000002687 Venezuelan Equine Encephalomyelitis Diseases 0.000 description 1
- 201000009145 Venezuelan equine encephalitis Diseases 0.000 description 1
- 201000009693 Venezuelan hemorrhagic fever Diseases 0.000 description 1
- 210000003501 Vero Cells Anatomy 0.000 description 1
- 241000607272 Vibrio parahaemolyticus Species 0.000 description 1
- 241000607265 Vibrio vulnificus Species 0.000 description 1
- 241001416177 Vicugna pacos Species 0.000 description 1
- 208000009421 Viral Pneumonia Diseases 0.000 description 1
- 206010047461 Viral infection Diseases 0.000 description 1
- 206010047504 Visceral larva migran Diseases 0.000 description 1
- 206010047741 Vulval cancer Diseases 0.000 description 1
- 206010047802 Waldenstrom's macroglobulinaemias Diseases 0.000 description 1
- 208000007510 West Nile Fever Diseases 0.000 description 1
- 241000244005 Wuchereria bancrofti Species 0.000 description 1
- 208000003152 Yellow Fever Diseases 0.000 description 1
- 241000607479 Yersinia pestis Species 0.000 description 1
- 241000607477 Yersinia pseudotuberculosis Species 0.000 description 1
- 208000001455 Zika Virus Infection Diseases 0.000 description 1
- 201000004296 Zika fever Diseases 0.000 description 1
- 239000003070 absorption delaying agent Substances 0.000 description 1
- 238000006640 acetylation reaction Methods 0.000 description 1
- 230000002378 acidificating Effects 0.000 description 1
- 201000007691 actinomycosis Diseases 0.000 description 1
- 239000012082 adaptor molecule Substances 0.000 description 1
- 239000011543 agarose gel Substances 0.000 description 1
- 235000004279 alanine Nutrition 0.000 description 1
- 150000001412 amines Chemical class 0.000 description 1
- 238000004458 analytical method Methods 0.000 description 1
- 201000010645 angiostrongyliasis Diseases 0.000 description 1
- 239000003242 anti bacterial agent Substances 0.000 description 1
- 238000009175 antibody therapy Methods 0.000 description 1
- 108010042982 antiglomerular basement membrane antibody Proteins 0.000 description 1
- 201000011165 anus cancer Diseases 0.000 description 1
- 125000003118 aryl group Chemical group 0.000 description 1
- 201000009361 ascariasis Diseases 0.000 description 1
- 235000009582 asparagine Nutrition 0.000 description 1
- 235000003704 aspartic acid Nutrition 0.000 description 1
- 201000002909 aspergillosis Diseases 0.000 description 1
- 230000001363 autoimmune Effects 0.000 description 1
- 230000003376 axonal Effects 0.000 description 1
- 201000008680 babesiosis Diseases 0.000 description 1
- 201000001178 bacterial pneumonia Diseases 0.000 description 1
- 239000003139 biocide Substances 0.000 description 1
- 229920000249 biocompatible polymer Polymers 0.000 description 1
- 238000006065 biodegradation reaction Methods 0.000 description 1
- 201000006824 bubonic plague Diseases 0.000 description 1
- 239000006189 buccal tablet Substances 0.000 description 1
- 239000001506 calcium phosphate Substances 0.000 description 1
- 229910000389 calcium phosphate Inorganic materials 0.000 description 1
- 235000011010 calcium phosphates Nutrition 0.000 description 1
- 201000004927 campylobacteriosis Diseases 0.000 description 1
- 201000003984 candidiasis Diseases 0.000 description 1
- 125000001314 canonical amino-acid group Chemical group 0.000 description 1
- 239000002775 capsule Substances 0.000 description 1
- 201000009030 carcinoma Diseases 0.000 description 1
- 230000000747 cardiac effect Effects 0.000 description 1
- 230000003915 cell function Effects 0.000 description 1
- 201000007455 central nervous system cancer Diseases 0.000 description 1
- 201000010353 central nervous system germ cell tumor Diseases 0.000 description 1
- 201000004308 chancroid Diseases 0.000 description 1
- 125000003636 chemical group Chemical group 0.000 description 1
- 201000006082 chickenpox Diseases 0.000 description 1
- 201000009182 chikungunya Diseases 0.000 description 1
- 229940048961 cholinesterase Drugs 0.000 description 1
- 201000009047 chordoma Diseases 0.000 description 1
- 238000004587 chromatography analysis Methods 0.000 description 1
- 230000035512 clearance Effects 0.000 description 1
- 238000010367 cloning Methods 0.000 description 1
- 239000011248 coating agent Substances 0.000 description 1
- 238000000576 coating method Methods 0.000 description 1
- 201000003486 coccidioidomycosis Diseases 0.000 description 1
- 229920001436 collagen Polymers 0.000 description 1
- 229960005188 collagen Drugs 0.000 description 1
- 201000011231 colorectal cancer Diseases 0.000 description 1
- 229910052802 copper Inorganic materials 0.000 description 1
- 239000010949 copper Substances 0.000 description 1
- RYGMFSIKBFXOCR-UHFFFAOYSA-N copper Chemical compound [Cu] RYGMFSIKBFXOCR-UHFFFAOYSA-N 0.000 description 1
- 201000007336 cryptococcosis Diseases 0.000 description 1
- VOLSCWDWGMWXGO-UHFFFAOYSA-N cyclobuten-1-yl acetate Chemical compound CC(=O)OC1=CCC1 VOLSCWDWGMWXGO-UHFFFAOYSA-N 0.000 description 1
- 201000002641 cyclosporiasis Diseases 0.000 description 1
- 235000018417 cysteine Nutrition 0.000 description 1
- 201000000077 cysticercosis Diseases 0.000 description 1
- 201000008167 cystoisosporiasis Diseases 0.000 description 1
- 230000001086 cytosolic Effects 0.000 description 1
- 210000001151 cytotoxic T lymphocyte Anatomy 0.000 description 1
- 125000001295 dansyl group Chemical group [H]C1=C([H])C(N(C([H])([H])[H])C([H])([H])[H])=C2C([H])=C([H])C([H])=C(C2=C1[H])S(*)(=O)=O 0.000 description 1
- 230000001934 delay Effects 0.000 description 1
- 201000002949 dengue disease Diseases 0.000 description 1
- 230000001419 dependent Effects 0.000 description 1
- 230000001627 detrimental Effects 0.000 description 1
- 201000004587 dientamoebiasis Diseases 0.000 description 1
- 235000005911 diet Nutrition 0.000 description 1
- 230000037213 diet Effects 0.000 description 1
- 238000010494 dissociation reaction Methods 0.000 description 1
- 230000005593 dissociations Effects 0.000 description 1
- 150000002019 disulfides Chemical class 0.000 description 1
- 241001493065 dsRNA viruses Species 0.000 description 1
- 201000005901 endemic typhus Diseases 0.000 description 1
- 230000002357 endometrial Effects 0.000 description 1
- 239000003623 enhancer Substances 0.000 description 1
- 230000002708 enhancing Effects 0.000 description 1
- 201000004101 esophageal cancer Diseases 0.000 description 1
- 239000003797 essential amino acid Substances 0.000 description 1
- 235000020776 essential amino acid Nutrition 0.000 description 1
- 239000005038 ethylene vinyl acetate Substances 0.000 description 1
- OGPBJKLSAFTDLK-UHFFFAOYSA-N europium Chemical compound [Eu] OGPBJKLSAFTDLK-UHFFFAOYSA-N 0.000 description 1
- 201000005884 exanthem Diseases 0.000 description 1
- 201000005866 exanthema subitum Diseases 0.000 description 1
- 201000001342 fallopian tube cancer Diseases 0.000 description 1
- 238000009472 formulation Methods 0.000 description 1
- 125000002485 formyl group Chemical group [H]C(*)=O 0.000 description 1
- 201000010175 gallbladder cancer Diseases 0.000 description 1
- 229910052733 gallium Inorganic materials 0.000 description 1
- 239000007789 gas Substances 0.000 description 1
- 201000000628 gas gangrene Diseases 0.000 description 1
- 230000002496 gastric Effects 0.000 description 1
- 238000001502 gel electrophoresis Methods 0.000 description 1
- 229920000159 gelatin Polymers 0.000 description 1
- 239000008273 gelatin Substances 0.000 description 1
- 239000007903 gelatin capsule Substances 0.000 description 1
- 235000019322 gelatine Nutrition 0.000 description 1
- 235000011852 gelatine desserts Nutrition 0.000 description 1
- 201000003950 geotrichosis Diseases 0.000 description 1
- 201000006592 giardiasis Diseases 0.000 description 1
- 235000013922 glutamic acid Nutrition 0.000 description 1
- 239000004220 glutamic acid Substances 0.000 description 1
- 201000000128 gnathomiasis Diseases 0.000 description 1
- 201000010536 head and neck cancer Diseases 0.000 description 1
- 230000036541 health Effects 0.000 description 1
- 229910001385 heavy metal Inorganic materials 0.000 description 1
- 201000005787 hematologic cancer Diseases 0.000 description 1
- 201000010284 hepatitis E Diseases 0.000 description 1
- 201000002563 histoplasmosis Diseases 0.000 description 1
- 201000009162 human monocytic ehrlichiosis Diseases 0.000 description 1
- 230000002209 hydrophobic Effects 0.000 description 1
- 230000001900 immune effect Effects 0.000 description 1
- 238000009169 immunotherapy Methods 0.000 description 1
- 239000007943 implant Substances 0.000 description 1
- 238000000099 in vitro assay Methods 0.000 description 1
- 238000011065 in-situ storage Methods 0.000 description 1
- 230000002779 inactivation Effects 0.000 description 1
- 238000010348 incorporation Methods 0.000 description 1
- 239000003701 inert diluent Substances 0.000 description 1
- 201000011422 infant botulism Diseases 0.000 description 1
- 201000006747 infectious mononucleosis Diseases 0.000 description 1
- 230000002757 inflammatory Effects 0.000 description 1
- 200000000018 inflammatory disease Diseases 0.000 description 1
- 239000007972 injectable composition Substances 0.000 description 1
- 229940079866 intestinal antibiotics Drugs 0.000 description 1
- 238000001361 intraarterial administration Methods 0.000 description 1
- 201000008893 intraocular retinoblastoma Diseases 0.000 description 1
- 238000007913 intrathecal administration Methods 0.000 description 1
- 238000010253 intravenous injection Methods 0.000 description 1
- 239000007951 isotonicity adjuster Substances 0.000 description 1
- 201000010982 kidney cancer Diseases 0.000 description 1
- 230000003902 lesions Effects 0.000 description 1
- 150000002632 lipids Chemical class 0.000 description 1
- 239000008297 liquid dosage form Substances 0.000 description 1
- 239000006193 liquid solution Substances 0.000 description 1
- 201000007270 liver cancer Diseases 0.000 description 1
- 201000005202 lung cancer Diseases 0.000 description 1
- 230000001926 lymphatic Effects 0.000 description 1
- 201000007919 lymphoplasmacytic lymphoma Diseases 0.000 description 1
- 239000012139 lysis buffer Substances 0.000 description 1
- 201000004792 malaria Diseases 0.000 description 1
- 201000003175 male breast cancer Diseases 0.000 description 1
- 230000003211 malignant Effects 0.000 description 1
- 201000005505 measles Diseases 0.000 description 1
- 201000004015 melioidosis Diseases 0.000 description 1
- 201000009906 meningitis Diseases 0.000 description 1
- 201000011475 meningoencephalitis Diseases 0.000 description 1
- 201000001198 metagonimiasis Diseases 0.000 description 1
- 230000001394 metastastic Effects 0.000 description 1
- VKQFCGNPDRICFG-UHFFFAOYSA-N methyl 2-methylpropyl 2,6-dimethyl-4-(2-nitrophenyl)-1,4-dihydropyridine-3,5-dicarboxylate Chemical compound COC(=O)C1=C(C)NC(C)=C(C(=O)OCC(C)C)C1C1=CC=CC=C1[N+]([O-])=O VKQFCGNPDRICFG-UHFFFAOYSA-N 0.000 description 1
- 238000007069 methylation reaction Methods 0.000 description 1
- 239000004530 micro-emulsion Substances 0.000 description 1
- 201000000090 microsporidiosis Diseases 0.000 description 1
- 201000009251 multiple myeloma Diseases 0.000 description 1
- 230000035772 mutation Effects 0.000 description 1
- 201000003793 myelodysplastic syndrome Diseases 0.000 description 1
- 230000002071 myeloproliferative Effects 0.000 description 1
- 229950006238 nadide Drugs 0.000 description 1
- 201000009240 nasopharyngitis Diseases 0.000 description 1
- 201000008026 nephroblastoma Diseases 0.000 description 1
- 230000001537 neural Effects 0.000 description 1
- 230000003472 neutralizing Effects 0.000 description 1
- 238000007899 nucleic acid hybridization Methods 0.000 description 1
- 201000008859 olfactory neuroblastoma Diseases 0.000 description 1
- 229940005935 ophthalmologic Antibiotics Drugs 0.000 description 1
- 201000000901 ornithosis Diseases 0.000 description 1
- 201000006958 oropharynx cancer Diseases 0.000 description 1
- 230000002611 ovarian Effects 0.000 description 1
- 201000002528 pancreatic cancer Diseases 0.000 description 1
- 201000000301 paracoccidioidomycosis Diseases 0.000 description 1
- 201000007052 paranasal sinus cancer Diseases 0.000 description 1
- 230000000849 parathyroid Effects 0.000 description 1
- 238000007911 parenteral administration Methods 0.000 description 1
- 230000000275 pharmacokinetic Effects 0.000 description 1
- 201000008006 pharynx cancer Diseases 0.000 description 1
- 201000008199 pleuropulmonary blastoma Diseases 0.000 description 1
- 229920001200 poly(ethylene-vinyl acetate) Polymers 0.000 description 1
- 229920000747 poly(lactic acid) polymer Polymers 0.000 description 1
- 239000004633 polyglycolic acid Substances 0.000 description 1
- 239000004626 polylactic acid Substances 0.000 description 1
- 230000002516 postimmunization Effects 0.000 description 1
- 238000001556 precipitation Methods 0.000 description 1
- 230000003449 preventive Effects 0.000 description 1
- 108090000765 processed proteins & peptides Proteins 0.000 description 1
- 102000004196 processed proteins & peptides Human genes 0.000 description 1
- 230000002035 prolonged Effects 0.000 description 1
- 230000004223 radioprotective Effects 0.000 description 1
- 239000011535 reaction buffer Substances 0.000 description 1
- 238000010188 recombinant method Methods 0.000 description 1
- 201000001275 rectum cancer Diseases 0.000 description 1
- 230000000306 recurrent Effects 0.000 description 1
- 210000003289 regulatory T cell Anatomy 0.000 description 1
- 230000000717 retained Effects 0.000 description 1
- 230000001177 retroviral Effects 0.000 description 1
- 201000004282 rickettsialpox Diseases 0.000 description 1
- 201000005404 rubella Diseases 0.000 description 1
- 150000003839 salts Chemical class 0.000 description 1
- 239000000523 sample Substances 0.000 description 1
- 201000004409 schistosomiasis Diseases 0.000 description 1
- 239000008299 semisolid dosage form Substances 0.000 description 1
- 230000035945 sensitivity Effects 0.000 description 1
- 201000003176 severe acute respiratory syndrome Diseases 0.000 description 1
- 231100000486 side effect Toxicity 0.000 description 1
- 230000019491 signal transduction Effects 0.000 description 1
- 230000011664 signaling Effects 0.000 description 1
- 238000001542 size-exclusion chromatography Methods 0.000 description 1
- 201000002612 sleeping sickness Diseases 0.000 description 1
- 238000010583 slow cooling Methods 0.000 description 1
- KEAYESYHFKHZAL-UHFFFAOYSA-N sodium Chemical compound [Na] KEAYESYHFKHZAL-UHFFFAOYSA-N 0.000 description 1
- 239000011734 sodium Substances 0.000 description 1
- 229910052708 sodium Inorganic materials 0.000 description 1
- 239000001509 sodium citrate Substances 0.000 description 1
- 239000007909 solid dosage form Substances 0.000 description 1
- 201000002190 staphyloenterotoxemia Diseases 0.000 description 1
- 230000001954 sterilising Effects 0.000 description 1
- 238000004659 sterilization and disinfection Methods 0.000 description 1
- 238000003860 storage Methods 0.000 description 1
- 238000007920 subcutaneous administration Methods 0.000 description 1
- 239000000829 suppository Substances 0.000 description 1
- 239000004094 surface-active agent Substances 0.000 description 1
- 230000002459 sustained Effects 0.000 description 1
- 230000002522 swelling Effects 0.000 description 1
- 201000010874 syndrome Diseases 0.000 description 1
- 239000006188 syrup Substances 0.000 description 1
- 235000020357 syrup Nutrition 0.000 description 1
- 230000002381 testicular Effects 0.000 description 1
- 201000003120 testicular cancer Diseases 0.000 description 1
- 201000009365 thymic carcinoma Diseases 0.000 description 1
- 201000002510 thyroid cancer Diseases 0.000 description 1
- 201000009642 tinea barbae Diseases 0.000 description 1
- 201000003875 tinea corporis Diseases 0.000 description 1
- 201000005882 tinea unguium Diseases 0.000 description 1
- 230000000699 topical Effects 0.000 description 1
- 230000002588 toxic Effects 0.000 description 1
- 201000005485 toxoplasmosis Diseases 0.000 description 1
- 102000003995 transcription factors Human genes 0.000 description 1
- 108090000464 transcription factors Proteins 0.000 description 1
- 230000002103 transcriptional Effects 0.000 description 1
- 238000002054 transplantation Methods 0.000 description 1
- 201000007588 trichinosis Diseases 0.000 description 1
- 239000011778 trisodium citrate Substances 0.000 description 1
- 230000001573 trophoblastic Effects 0.000 description 1
- 201000008827 tuberculosis Diseases 0.000 description 1
- 201000008297 typhoid fever Diseases 0.000 description 1
- 241000701161 unidentified adenovirus Species 0.000 description 1
- 241000701447 unidentified baculovirus Species 0.000 description 1
- 241001430294 unidentified retrovirus Species 0.000 description 1
- 201000005112 urinary bladder cancer Diseases 0.000 description 1
- 238000002255 vaccination Methods 0.000 description 1
- 238000001291 vacuum drying Methods 0.000 description 1
- 238000009777 vacuum freeze-drying Methods 0.000 description 1
- 201000001042 variant Creutzfeldt-Jakob disease Diseases 0.000 description 1
- 201000011528 vascular disease Diseases 0.000 description 1
- 230000017613 viral reproduction Effects 0.000 description 1
- 235000012431 wafers Nutrition 0.000 description 1
- 238000005406 washing Methods 0.000 description 1
- VWQVUPCCIRVNHF-OUBTZVSYSA-N yttrium-90 Chemical compound [90Y] VWQVUPCCIRVNHF-OUBTZVSYSA-N 0.000 description 1
Abstract
Polypeptides, such as antibody molecules and TCR molecules, and methods of making the same, are disclosed. The polypeptides can be used to treat, prevent, and/or diagnose disorders.
Description
BINDING POLYPEPTIDES AND METHODS OF MAKING THE SAME
CROSS REFERENCE TO RELATED APPLICATIONS
This application claims the benefit of US. ional Application No. 62/438,712, filed
December 23, 2016. The contents of the aforesaid application are hereby incorporated by reference in
their entirety.
BACKGROUND
Monoclonal antibody ies are a class of immunotherapies that involve monoclonal
antibodies (mAbs) that are capable of specifically interacting with disease-relevant biological
molecules. In recent years, the disease areas that therapeutic antibodies can target have significantly
expanded, and a number of monoclonal antibodies and antibody-derivative products have been
ed for therapeutic use in the United States and many other countries. Monoclonal antibody
therapies are currently used or investigated for treating various diseases or conditions, including, for
example, infectious diseases, cancer, immune diseases, organ transplantation, vascular diseases,
and metabolic diseases.
Given the ability of monoclonal antibodies and antibody-derivative products in modulating
various biological functions, the need exists for developing new approaches for generation of
antibodies suitable for treating, preventing, and diagnosing disorders.
SUMMARY
This disclosure provides, at least in part, binding polypeptides (e.g., dy molecules or T-
cell receptor (TCR) molecules) that comprise one or more of the structural or functional properties
sed herein. In an embodiment, libraries of the binding polypeptides, s for making the
ptides or libraries, nucleic acid molecules encoding the binding polypeptides, expression
vectors, host cells, compositions (e.g., pharmaceutical compositions), kits, and containers, are also
provided. The polypeptides (e.g., antibody les or TCR molecules) disclosed herein can be
used (alone or in combination with other agents or therapeutic modalities) to treat, prevent and/or
diagnose ers, such as disorders and conditions disclosed herein.
In an , the disclosure features a method of making a nucleic acid ce comprising a
sequence that s a heavy chain element (HC element) of an antibody heavy chain variable
region (HCVR) and a light chain element (LC element) of an antibody light chain variable region
(LCVR), and wherein the HCVR and LCVR are matched, the method comprising:
a) acquiring an isolated production reaction site, e.g., a production micro-chamber,
comprising:
WO 19402
i) a heavy chain (HC) strand, wherein the HC strand is a strand of a heavy chain double-
stranded cDNA (HC ds cDNA) comprising a segment that encodes an HC element of the HCVR from
a cell, e. g., a heavy chain variable region sequence (HCVRS); and
ii) a light chain (LC) strand, wherein the LC strand is a strand of a light chain double-stranded
cDNA (LC ds cDNA) comprising a segment that encodes an LC element of the LCVR from the cell,
e.g., a light chain variable region sequence ), and
b) covalent linking, e. g., ligation, of an HC strand to an LC strand,
wherein the isolated production reaction site, e.g., a production micro-chamber, does not
include a nucleic acid encoding an HCVR or an LCVR from a cell other than the cell (e. g., a ent
cell, e.g., a different B cell),
thereby making a nucleic acid sequence comprising a sequence that encodes an HC element
of an HCVR and a LC element of an LCVR, wherein the HCVR and LCVR are matched.
In an embodiment, the HC element comprises, or consists of, an HCVRS, or a functional
fragment thereof (e.g., an antigen binding fragment thereof). In an embodiment, the LC element
comprises, or ts of, an LCVRS, or a functional nt thereof (e.g., an antigen binding
nt thereof).
In an ment, the HC ds cDNA comprises a segment that encodes an HCVRS. In an
embodiment, the LC ds cDNA comprises a segment that encodes an LCVRS. In an embodiment, the
HC ds cDNA comprises a segment that encodes an HCVRS, and the LC ds cDNA comprises a
segment that encodes an LCVRS.
In an embodiment, the cell is an immune cell, e.g., a B cell, e.g., a human B cell. In an
embodiment, the cell is a mammalian cell or an avian cell.
In an embodiment, the nucleic acid ce is configured such that, when expressed, the HC
element and the LC element (6.3., the HCVRS and the LCVRS) form a functional antigen binding
molecule, e.g., an scFv, an Fab, or an scFab. In an embodiment, the antigen binding molecule, e.g.,
an scFv, is functional in vitro, ex vivo, or in vivo, e.g., as determined by a method or assay bed
In an embodiment, acquiring an isolated production reaction site, e.g., a production micro-
chamber, comprises:
a) acquiring a capture substrate bound to: (i) a first double-stranded cDNA (ds cDNA)
comprising a strand that is complementary to a first mRNA that encodes an HCVR from a cell; and
(ii) a second ds cDNA sing a strand complementary to a second mRNA encoding an LCVR
from the cell (the cDNA loaded capture substrate), and
b) maintaining the ed production reaction site, e.g., the production micro-chamber,
under conditions that allow amplification of the first and second ds cDNAs, to e: a plurality of
HC ds cDNAs comprising a segment that encodes an HC element of the HCVR from the cell, e.g., an
HCVRS; and a plurality of LC ds cDNAs comprising a segment that encodes an LC element of the
LCVR from the cell, e. g., an LCVRS.
In an embodiment, the HC ds cDNA is identical, or substantially identical, to the first ds
cDNA. For example, the sense strand of the HC ds cDNA is at least 80%, 85 %, 90%, 95%, 98%,
99%, or 100% cal to, or differs by no more than 1, 2, 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50
nucleotides from, the sense strand of the first ds cDNA, and/or the antisense strand of the HC ds
cDNA is at least 80%, 85%, 90%, 95%, 98%, 99%, or 100% identical to, or differs by no more than 1,
2, 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50 nucleotides from, the antisense strand of the first ds cDNA.
In an embodiment, the LC ds cDNA is identical, or substantially identical, to the second ds
cDNA. For example, the sense strand of the LC ds cDNA is at least 80%, 85%, 90%, 95%, 98%,
99%, or 100% cal to, or differs by no more than 1, 2, 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50
nucleotides from, the sense strand of the second ds cDNA, and/or the antisense strand of the LC ds
cDNA is at least 80%, 85%, 90%, 95%, 98%, 99%, or 100% identical to, or differs by no more than 1,
2, 5, 10, 15, 20, 25, 30, 35, 40, 45 , or 50 nucleotides from, the antisense strand of the second ds
cDNA.
In an embodiment, the HC strand is a sense strand. In an embodiment, the LC strand is a
sense strand. In an embodiment, the HC strand is an antisense strand. In an embodiment, the LC
strand is an antisense strand. In an embodiment, both the HC strand and the LC strand are sense
strands. In an embodiment, both the HC strand and the LC strand are antisense strands.
In an embodiment, the capture ate comprises a bead, e.g., a magnetic bead. In an
ment, the e substrate comprises a moiety (e.g., an oligonucleotide) which binds to
cDNA, e.g., (i) a moiety which binds to the HC ; (ii) a moiety which binds to the LC strand; or
(iii) both (i) and (ii). In an embodiment, the moiety which binds to the HC strand is different from the
moiety which binds to the LC strand, e.g., to facilitate creating conditions favorable to capturing
similar levels of each DNA molecule type. In an embodiment, the moiety which binds to the HC
strand is identical to the moiety which binds to the LC strand.
In an embodiment, the first mRNA and the second mRNA are disposed on an mRNA loaded
capture substrate.
In an embodiment, the isolated production reaction site, e.g., the production chamber,
comprises: a t mixture suitable for producing, from the first and second mRNAs (e.g., after the
first and second mRNAs are released from the mRNA loaded capture substrate into a solution), a first
ds cDNA comprising a segment that encodes an HC element of the HCVR of the cell, e.g., an
HCVRS, and a second ds cDNA comprising a segment that encodes an LC element of the LCVR of
the cell, e.g., an LCVRS.
In an embodiment, the isolated production reaction site, e.g., production micro-chamber,
comprises primers that mediate the tion of the first ds cDNA. In an embodiment, the isolated
production reaction site, e.g., tion micro-chamber, comprises primers that mediate the
production of the second ds cDNA.
In an embodiment, a cDNA strand that is complementary to a first mRNA that encodes an
HCVR from a cell is made by reverse transcription of the first mRNA. In an embodiment, a cDNA
strand that is complementary to a second mRNA that encodes an LCVR from a cell is made by
reverse transcription of the second mRNA.
In an embodiment, the e transcription takes place in the isolated production reaction
site, e.g., a production-micro chamber. In an embodiment, the reverse transcription takes place in an
isolated cell reaction site, e.g., a cell isolation micro-chamber. In an embodiment, the e
transcription takes place outside the isolated production reaction site, e.g., a production micro-
chamber, or outside an isolated cell reaction site, e.g., a cell isolation micro-chamber. In an
embodiment, the reverse transcription takes place outside the isolated production reaction site, e.g., a
production-micro chamber, and outside an isolated cell on site, e.g., a cell isolation micro-
chamber. In an embodiment, the e ription takes place outside an isolated reaction site,
e.g., outside a micro-chamber.
In an ment, the amplification comprises 30 or fewer cycles, e.g., 20 or fewer cycles,
e.g., 15 or fewer, 14 or fewer, 13 or fewer, 12 or fewer, 11 or fewer, 10 or fewer, 9 or fewer, 8 or
fewer, 7 or fewer, 6 or fewer, or 5 or fewer cycles.
In an embodiment, the reverse transcription and/or amplification uses one or more primers,
e.g., comprising a sequence specific for an HCVRS and/or an LCVRS.
In an embodiment, the reverse transcription and/or amplification comprises using two or more
primers that mediate the production of the HC ds cDNA, wherein at least one primer comprises a
nucleotide modification, and wherein at least one primer does not comprise a nucleotide modification.
In an embodiment, the amplification comprises using two or more primers that mediate the production
of the LC ds cDNA, wherein at least one primer comprises a nucleotide modification, and wherein at
least one primer does not se a tide modification.
In an ment, at least one primer comprises a nucleotide modification, e.g., which
reduces, e.g., inhibits, DNA synthesis, 6.3., by a DNA polymerase. In an embodiment, at least one
primer does not comprise a tide modification, e.g., which reduces, e.g., inhibits, DNA
synthesis, e.g., by a DNA polymerase.
In an embodiment, the nucleotide modification inhibits a DNA polymerase from extending
the DNA. Without wishing to be bound by theory, it is believed that in an ment, any chemical
entity that reduces (e.3., blocks) DNA rase extension can be used in accordance with the
methods described herein.
In an embodiment, the nucleotide modification is an insertion of a spacer to the primer, e. g.,
n two adjacent nucleotides in the primer. In an embodiment, the spacer is a flexible spacer. In
an embodiment, the spacer is a carbon spacer (e.g., -(CH2)n-, wherein n=3, 4, 5, 6, 7, 8, 9, 10, or
WO 19402
more), two or more (e.g., three, four, five, six, seven, eight, nine, ten, or more) abasic nucleotides, or a
polyethylene glycol (PEG) spacer. In an embodiment, the spacer is a PEG spacer. In an embodiment,
the nucleotide modification is 2’-O-methyl, 2’-OH, , or uracil, e.g., to a ribose.
In an embodiment, the nucleotide modification is located internally or at the 3’ end of the
primer. In an embodiment, at least one primer comprises (i) a first member; (ii) a second member;
and optionally (iii) a third member, e.g., comprising a nucleotide modification described herein, e.g.,
located between (i) and (ii).
In an embodiment, the first member is capable of ing with the second member. In an
embodiment, the first member is capable of annealing with the second member in the same primer,
e.g., through intra-molecular hybridization, e.g., to form a hairpin structure comprising a duplex
region of 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, more basepairs. In another
embodiment, the first member is capable of ing hybridizing with the second member in a
different primer, e.g., through inter-molecular hybridization, e.g., to form a double-stranded ure
comprising a duplex region of 4, 5,6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, more
basepairs. Without wishing to be bound by theory, it is believed that in an embodiment, there are at
least two secondary structures that the modified primers can form and facilitate reduction (e.g.,
tion) of competition to substrate (e.g., bead) capture. For example, the secondary structure can
be a hairpin-like structure formed by intra-molecular hybridization (within the same primer), or the
secondary structure can be a duplex structure formed by inter-molecular hybridization (between two
ent primers).
In an embodiment, the first member comprises a ce that is complementary to the
sequence of an oligonucleotide attached to the capture substrate. In an embodiment, the second
member comprises (e.g., from 5’ to 3’) one, two, or all of: (i) a sequence that is complementary to at
least a portion of the first member; (ii) a universal priming sequence (e.g., for PCR amplification or
next-generation cing); and (iii) a sequence complementary to a target sequence, e.g., an
HCVRS and/or an LCVRS. In an embodiment, the universal priming sequence is identical, or
substantially identical, to the sequence that is complementary to at least a portion of the first .
In r embodiment, the universal priming sequence is different from the sequence that is
mentary to at least a portion of the first member. In an embodiment, the second member
comprises a sequence for gous recombination (e.g., in a yeast or mammalian cell).
In an embodiment, at least one primer comprises a ce encoding at least a portion of a
linker sequence, or a complementary sequence thereof. In an embodiment, the primer that comprises
a sequence encoding at least a portion of a linker sequence, or a complementary sequence thereof, is
phosphorylated, e.g., 5’ phosphorylated. Without wishing to be bound by theory, it is believed that in
an ment, any sequence with the general properties of flexibility (e.g., tated by glycine)
and hydrophilicity can work effectively in accordance with the methods described herein. ary
s can generally have overrepresentation of one or more of Gly, Ser, Thr, or Ala and
underrepresentation of hydrophobic residues, e.g., one or more of Trp, Tyr, Phe, Cys, Met, Leu, or Ile.
The length of the primer may vary, e.g., 3-50 amino acid residues (e.g., 5-45, 10-40, 15-35, 20-30, 10-
, 10-30, 20-40, or 30-40 amino acid residues). In an embodiment, the linker sequence comprises, or
consists of, ((Gly)m-Ser))n, where m=3, 4, 5, or more and n=1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more. In an
embodiment, the linker sequence comprises, or consists of, ly-Gly-Gly-Ser)n, where n=1, 2, 3,
4, 5, 6, 7, 8, 9, 10, or more.
In an embodiment, the primer is a primer described herein, e.g., in Examples.
In an embodiment, the reverse transcription, the amplification, or both, occurs in a solution in
the isolated production reaction site, e.g., production chamber. In an embodiment, the e
transcription, the amplification, or both, does not occur on the ate (e.g., bead). For example, the
reverse transcription, the amplification, or both, can occur on in a solution within a droplet.
In an embodiment, the HC ds cDNA comprises a 5’ overhang, e.g., a 5’ overhang that is
e of hybridizing to an oligonucleotide attached to a capture substrate. In an ment, the
HC ds cDNA ses a blunt end, e.g., a blunt end comprising a 5’ phosphate. In an ment,
the LC ds cDNA comprises a 5’ overhang, e.g., a 5’ overhang that is capable of hybridizing to an
oligonucleotide attached to a capture substrate. In an embodiment, the LC ds cDNA comprises a
blunt end, e.g., a blunt end comprising a 5’ phosphate. In an embodiment, the HC ds cDNA and the
LC ds cDNA comprise sticky ends, e.g., both have 5 ’ overhangs.
In an embodiment, the HC strand and the LC strand are covalently linked, e.g., ligated, to
produce a single stranded nucleic acid sequence, wherein the HC and LC strands are both sense
strands or both antisense strands. In an embodiment, a denatured HC strand of the HC ds cDNA to a
red LC strand of the LC ds cDNA are covalently linked, e.g., ligated, wherein the HC and LC
strands are both sense strands or both antisense strands. In an embodiment, the HC strand is present
in the HC ds cDNA and the LC strand is present in the LC ds cDNA, and wherein the HC ds cDNA
and the LC ds cDNA are covalently linked, e.g., d, e.g., to produce a double stranded nucleic
acid sequence.
In an embodiment, the covalent linking, e. g., ligation, occurs in the isolated production
reaction site. In an embodiment, the isolated production reaction site, e.g., a production micro-
chamber, or the isolated linkage reaction site, e.g., a linkage micro-chamber, comprises a reagent that
is e of covalently linking, e.g., ligating, the HC and LC strands or the HC and LC ds cDNAs.
In an embodiment, the ed production reaction site, e.g., a production micro-chamber comprises
an enzyme that covalently couples the HC and LC strands or the HC and LC ds cDNAs. In an
embodiment, the enzyme is a ligase, e. g., a thermal stable ligase. In an embodiment, the covalent
linking comprises ligase thermocycling.
In an ment, the covalent linking, e. g., ligation, occurs in a site ent from the
ed production reaction site, e.g., occurs in an isolated linkage reaction site, e.g., a linkage micro-
chamber. In an embodiment, the HC strand and the LC strand are transferred from the isolated
production site to the isolated linkage reaction site, e.g., a linkage micro-chamber, and the nt
linking occurs in the isolated linkage on site, e.g., a linkage micro-chamber. In an embodiment,
the ed linkage reaction site, e.g., a linkage micro-chamber, comprises a reagent that is capable of
covalently linking, e.g., ng, the HC and LC strands or the HC and LC ds cDNAs. In an
embodiment, the isolated linkage reaction site, e. g., a linkage micro-chamber, ses an enzyme
that ntly couples the HC and LC strands or the HC and LC ds cDNAs. In an embodiment, the
enzyme is a ligase, e.g., a thermal stable ligase. In an embodiment, the covalent linking comprises
ligase thermocycling.
In an embodiment, the covalent linking, e.g., ligation, comprises: (a) heating the isolated
linkage reaction site, e.g., the linkage micro-chamber, under conditions (e.g., at 950C) that allow
denaturation of the HC strand and the LC strand; (b) cooling the isolated linkage reaction site, e.g., the
linkage micro-chamber, under ions (e.g., at 50-65°C) that allow hybridization of the splint
oligonucleotide to the HC strand and the LC strand; (c) maintaining the isolated linkage reaction site,
e. g., the linkage micro-chamber, under conditions (e.g., at 45-65°C) that allow ligation of the HC
strand and the LC strand (e.g., formation of phosphodiester bond between the HC strand and the LC
strand); and (d) repeating steps (a), (b), and (c) sequentially for 2, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50,
or more cycles.
In an embodiment, the HC strand and the LC strand are covalently , e.g., d, in the
presence of a splint oligonucleotide. In an embodiment, the splint oligonucleotide is hybridized to a
ce comprising the junction of the HC strand and the LC strand, or a ce complementary
thereof, and forms a ed region at the site of ligation. In an embodiment, the splint
oligonucleotide comprises a modification (e.g., an NH; group) that inhibits DNA synthesis, e.g., by a
DNA polymerase. In an embodiment, the modification is at the 3’ end of the splint oligonucleotide.
In an embodiment, a strand complimentary to the covalently linked, e.g., ligated, HC and LC
strands is produced by amplification.
In an embodiment, the method, e.g., the step of covalent linkage, does not include a step of
p extension rase chain reaction (OE-PCR), also known as splicing by overlap ion
or splicing by overhang ion (SOE) PCR.
In an embodiment, the method further comprises, prior to acquiring the isolated production
reaction site, e.g., a production micro-chamber, acquiring an mRNA loaded capture substrate.
In an embodiment, acquiring the mRNA loaded capture substrate comprising: a) acquiring an
isolated cell reaction site, e.g., a cell ion micro-chamber, comprising: i) a cell; and ii) a capture
substrate capable of binding a first mRNA encoding an HCVR from the cell and a second mRNA
encoding an LCVR from the cell; and b) maintaining the isolated cell reaction site, e.g., the cell
isolation chamber, under conditions that allow lysis of the cell and binding of the capture
substrate with the first mRNA and the second mRNA to form the mRNA loaded capture substrate,
wherein the ed cell reaction site, e.g., cell isolation micro-chamber, does not include a c
acid encoding an HCVR or an LCVR from a cell other than the cell (e.g., a different cell).
In an embodiment, the isolated cell reaction site, e.g., cell isolation chamber, comprises
a lysing reagent, e.g., a detergent. In an embodiment, the cell is lysed by heat or an enzyme. In an
embodiment, the capture substrate comprises a moiety (e.g., an oligonucleotide) which binds mRNA,
6.3., an oligo(dT).
In an embodiment, the method further comprises releasing the mRNA loaded capture
substrate from the isolated cell reaction site, e.g., the cell isolation micro-chamber. In an
embodiment, the releasing step is performed in the presence of a poly(dA) or poly(dT)
oligonucleotide, e.g., to reduce cross-binding of non-captured mRNA.
In an embodiment, the mRNA loaded capture ate is transferred from the isolated cell
reaction site, e.g., the cell isolation micro-chamber, to the isolated production reaction site, e.g., the
production micro-chamber.
In an ment, the method further comprises releasing the nucleic acid ce from the
isolated production reaction site, e.g., the production chamber. In an embodiment, the method
further comprises amplifying the nucleic acid sequence. In an embodiment, amplification of the
nucleic acid sequence occurs outside the isolated production reaction site, e.g., the production micro-
chamber, e.g., after the nucleic acid is released from the isolated production reaction site, e.g., the
production micro-chamber. In an embodiment, amplification of the nucleic acid sequence occurs at
the isolated production on site, e. g., the production micro-chamber.
In an embodiment, the method r ses sequencing all or a portion of the nucleic
acid sequence.
In an embodiment, the method further comprises inserting all or a portion of nucleic acid
sequence into a vector. In an embodiment, the vector supplies an additional HC element or LC
element not included in the nucleic acid sequence. In an ment, the vector supplies an HC
CDRl, an HC CDRZ, or both. In an embodiment, the method further comprises expressing the
vector.
In an embodiment, the method further comprises expressing the nucleic acid sequence to
produce a polypeptide comprising a segment that encodes an HC element of the HCVR, e.g., an
HCVRS, and a segment that encodes an LC element of the LCVR, e.g., an LCVRS. In an
embodiment, the LC element is N-terminal to the HC element in the polypeptide. In an embodiment,
the HC element is C-terminal to the LC element in the polypeptide.
In an ment, the method r comprises contacting the polypeptide with an antigen.
In an embodiment, the method further comprises determining if the polypeptide binds the antigen, in
vitro, ex vivo, or in viva, e.g., by a method or assay bed herein.
In an aspect, the disclosure features a method of making a nucleic acid sequence comprising a
sequence that encodes a heavy chain element (HC element) of an antibody heavy chain variable
region (HCVR) and a light chain element (LC element) of an antibody light chain variable region
(LCVR), and wherein the HCVR and LCVR are matched, comprising:
a) acquiring an isolated cell reaction site (e.g., an isolated cell reaction site described herein),
6.3., a cell isolation micro-chamber, comprising: i) a cell (e.g., a cell described ); and ii) a
capture substrate (e. g., a capture substrate bed herein) capable of g a first mRNA
encoding an HCVR from the cell and a second mRNA encoding an LCVR from the cell;
b) maintaining the isolated cell reaction site, e.g., the cell isolation micro-chamber, under
conditions that allow lysis of the cell and binding of the capture substrate with the first mRNA and the
second mRNA to form an mRNA loaded capture substrate,
wherein the isolated cell reaction site, e.g., cell ion micro-chamber, does not include a
nucleic acid encoding an HCVR or an LCVR from a cell other than the cell (e.g., a different cell);
c) contacting the mRNA loaded e substrate with a reaction e, e.g., a reaction
mixture comprising reverse transcriptase, that uses the loaded mRNA as a template to make cDNA
(this can occur, e.g., in the isolated cell reaction site, in an isolated production reaction site, or in
neither, e.g., not in an isolated reaction site);
d) acquiring an ed production reaction site (e.g., an isolated production reaction site
described herein), e.g., a production chamber, comprising: i) a heavy chain (HC) strand,
wherein the HC strand is a strand of a heavy chain double-stranded cDNA (HC ds cDNA) comprising
a segment that encodes an HC element of the HCVR from the cell, e.g., a heavy chain variable region
sequence (HCVRS); and ii) a light chain (LC) strand, n the LC strand is a strand of a light
chain double-stranded cDNA (LC ds cDNA) comprising a t that encodes an LC element of the
LCVR from the cell, e.g., a light chain variable region sequence (LCVRS),
wherein the isolated production reaction site, e.g., a production micro-chamber, does not
include a nucleic acid encoding an LCVR or an HCVR from a cell other than the cell (e.g., a ent
cell); and
e) covalent linking, e.g., on, of the HC strand to the LC strand.
In an embodiment, one or more (e.g., two, three, four, or all) of the steps a)-e) are performed
in accordance with a method described herein. In an embodiment, each of the steps a)-e) is performed
in accordance with a method described herein.
In an aspect, the disclosure features a method of making a nucleic acid sequence sing a
sequence that encodes a heavy chain element (HC element) of an antibody heavy chain variable
region (HCVR) and a light chain element (LC element) of an antibody light chain variable region
(LCVR), and n the HCVR and LCVR are matched, comprising:
WO 19402
a) acquiring an ed cell reaction site (e.g., an isolated cell reaction site described herein),
e.g., a cell isolation micro-chamber, comprising: i) a cell (e.g., a cell described herein); and ii) a
capture substrate (e. g., a capture substrate described herein) capable of binding a first mRNA
encoding an HCVR from the cell and a second mRNA encoding an LCVR from the cell;
b) maintaining the ed cell reaction site, e.g., the cell isolation micro-chamber, under
conditions that allow lysis of the cell and binding of the capture substrate with the first mRNA and the
second mRNA to form the mRNA loaded capture substrate,
n the isolated cell reaction site, e.g., cell isolation micro-chamber, does not include a
nucleic acid encoding an HCVR or an LCVR from a cell other than the cell (e.g., a different cell);
c) acquiring an isolated production on site (e.g., an isolated production reaction site
bed herein), e.g., a production micro-chamber, comprises: contacting the mRNA loaded capture
substrate with a reaction mixture, e.g., a reaction mixture sing reverse transcriptase, that uses
the loaded mRNA as a template, to e: a first double-stranded cDNA (ds cDNA) comprising a
strand that is complementary to a first mRNA that encodes an HCVR from a cell; and a second ds
cDNA comprising a strand complementary to a second mRNA encoding an LCVR from the cell (the
cDNA loaded capture substrate);
n the isolated production reaction site, e.g., a production micro-chamber, does not
include a nucleic acid encoding an LCVR or an HCVR from a cell other than the cell (e. g., a different
cell).
d) maintaining the isolated tion reaction site, e.g., the production micro-chamber,
under conditions that allow amplification of the first and second ds cDNAs, to e: a plurality of
HC ds cDNAs comprising a segment that encodes an HC element of the HCVR from the cell, e. g., an
HCVRS; and a ity of LC ds cDNAs comprising a segment that encodes an LC element of the
LCVR from the cell, e.g., an LCVRS;
e) acquiring an ed linkage reaction site (e.g., an isolated linkage reaction site described
herein), e.g., a linkage micro-chamber, comprising: covalent linking, e.g., ligation, of a strand of the
HC ds cDNA (HC strand) to a strand of the LC ds cDNA (LC strand), wherein the HC and LC strands
are both sense strands or antisense strands; and
t) amplifying the ntly linked, e.g., ligated, HC and LC strands.
In an embodiment, one or more (e.g., two, three, four, five, or all) of the steps a)-f) are
performed in accordance with a method described herein. In an embodiment, each of the steps a)-f) is
performed in accordance with a method described herein.
In an aspect, the disclosure features a method of making a library comprising a plurality of
unique members, the method comprising:
making the ity of members, wherein each of the members comprises a ce that
encodes a heavy chain element (HC element) of a heavy chain variable region (HCVR) and a light
chain element (LC element) of a light chain le region (LCVR), and wherein the HCVR and
LCVR are matched, made by a method described herein,
wherein each unique nucleic acid sequence of the plurality comprises an HC element and an
LC element from a different unique cell (e.g., a cell described herein),
thereby making a y comprising a plurality of unique s.
In an embodiment, the plurality of unique members ses at least 104, 105, 106, 107, 108,
or 109 unique members. In an embodiment, the plurality of unique members comprises 104 to 109, 104
to 10“, 104 to 107, 104 to 106, 104 to 105, 108 to 109, 107 to 109, 106 to 109, 105 to 109, 105 to 108, 106 to
107, 104 to 105, 105 to 106, 106 to 107, 107 to 108, or 108 to 109 unique members. In an embodiment, at
least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%, of the members in the library are unique
members (which encode matched HC element and LC element sequences). In an ment, less
than 20%, 15%, 10%, 5%, 4%, 3%, 2%, or 1%, of the members in the library are unique s
(which encode matched HC element and LC element ces).
In an aspect, the disclosure features a library comprising a plurality of unique s,
wherein,
i) each unique member of the plurality comprises a segment that encodes an HC element, e.g.,
an HCVRS, and a segment that encodes an LC element, e. g., an LCVRS, wherein the HC element and
the LC element in each unique member is matched;
ii) each unique member of the plurality comprises a t that encodes an HC element,
6.3., an HCVRS, and a segment that encodes an LC element, e.g., an LCVRS, from a different unique
cell; and
iii) the library comprises one or more (e.g., two, three, four, or all) of the following
properties:
a) the library is made by a method described herein;
b) the plurality of unique s comprises at least 104, 105, 106, 107, 108, or 109
unique nucleic acid sequences;
c) the plurality of unique members comprises 104 to 109, 104 to 108, 104 to 107, 104 to
106, 104 to 105, 108 to 109, 107 to 109, 106 to 109, 105 to 109, 105 to 108, 106 to 107, 104 to 105,
105 to 106, 106 to 107, 107 to 108, or 108 to 109 unique members;
d) at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%, of the s in
the library are unique members (which encode matched HC element and LC element
sequences); or
e) less than 20%, 15%, 10%, 5%, 4%, 3%, 2%, or 1%, of the members in the library
are unique members (which encode matched HC element and LC element sequences).
In an embodiment, each unique member of the plurality is configured such that, when
expressed, the HC element, e.g., the HCVRS, and the LC element, e.g., the LCVRS, form a functional
antigen binding molecule, e.g., an scFv, an Fab, or an scFab.
In an embodiment, the library is a display y. In an embodiment, each of the members of
the plurality further encodes a ptide that results in display of the member on the surface of a
display entity. In an embodiment, the library is a phage y library. In an ment, the
library is a yeast display library. In an ment, the library is a ian y library.
In an aspect, the disclosure features a method of making a binding polypeptide (e.g., a
polypeptide comprising an HC element and an LC element), the method comprising: a) acquiring a
library described herein, e.g., by a method described ; and b) expressing a polypeptide encoded
by a unique nucleic acid of the y.
In an embodiment, the method further comprises contacting the polypeptide with an antigen.
In an embodiment, the method further comprises retrieving (e.g., isolating or purifying) the nucleic
acid that encodes a polypeptide that binds the antigen.
In an aspect, the disclosure features an isolated production reaction site, e.g., a production
micro-chamber, which is an ed production reaction site described herein (e.g., comprising a
nucleic acid encoding an HCVR and a nucleic acid encoding a LCVR, wherein the HCVR and the
LCVR are matched).
In an embodiment, the isolated production reaction site, e.g., a production micro-chamber,
does not include a nucleic acid encoding an HCVR or an LCVR from a different cell.
In an embodiment, the isolated production reaction site, e.g., a production micro-chamber,
comprises one, two, or all of: (i) one or more primers specific to V gene ces of the HC and LC;
(ii) one or more primers specific to overhangs introduced onto the HC and LC cDNAs; or (iii) one or
more s sing a first member, a second , and a third member comprising a
nucleotide modification (e.g., a spacer) located between the first and second members, wherein the
first member is capable of annealing with the second member of the same primer or a different
primer, e.g., forming a structure comprising a duplex region of 4, 5, 6, 7, 8, 9, 10, ll, 12, l3, 14, 15,
16, 17, 18, 19, 20, more basepairs.
In an embodiment, the ed production reaction site, e. g., a production micro-chamber,
does not comprise a reagent that can covalently link nucleic acids, e.g., a ligase, e.g., a thermostable
ligase. In another ment, the isolated production reaction site, e.g., a production micro-
chamber, comprises a reagent that can covalently link nucleic acids, e.g., a ligase, e.g., a thermostable
1i gase.
2017/068204
In an aspect, the sure features a self-annealing ucleotide comprising a first
member, a second member, and third member comprising a nucleotide modification (e.g., a spacer)
located between the first and second members, wherein the first member is capable of annealing with
the second member of the same ucleotide (e.g., for a method of making a nucleic acid sequence
comprising a sequence that encodes an HC element of an HCVR and a LC element of an LCVR,
wherein the HCVR and LCVR are matched).
In an embodiment, the first and second members are capable of forming a hairpin structure
comprising a duplex region of 4, 5,6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, more
basepairs. In an embodiment, the first member is 5-40 nucleotides, 6.3., 5-10, 5-20, 5-30, 30-40, 20—
40, 10-30, 10-30, or 15-25 nucleotides, in length. In an embodiment, the second member is 5-40
nucleotides, e.g., 5-10, 5-20, 5-30, 30-40, 20-40, 10-30, 10-30, or 15-25 nucleotides, in length.
In an embodiment, the spacer is a spacer described herein, e.g., a flexible spacer or a PEG
spacer.
In an embodiment, the first member comprises a sequence that is complementary to the
sequence of an oligonucleotide attached to a capture substrate.
In an ment, the second member comprises (e.g., from 5’ to 3’) one, two, or all of: (i) a
sequence that is mentary to at least a portion of the first member; (ii) a universal priming
sequence (e.g., for PCR amplification or next-generation sequencing); and (iii) a sequence
complementary to a target sequence, e.g., an HCVRS and/or an LCVRS. In an embodiment, the
universal priming sequence is identical, or ntially identical, to the sequence that is
mentary to at least a n of the first member. In another embodiment, the universal
priming sequence is different from the sequence that is mentary to at least a portion of the first
member. In an embodiment, the second member comprises a sequence for homologous
recombination (e.g., in a yeast or mammalian cell).
In an , the disclosure features an isolated linkage reaction site, e. g., a linkage micro-
chamber, which is an ed linkage reaction site described herein (e.g., comprising a nucleic acid
encoding an HCVR and a nucleic acid encoding a LCVR, wherein the HCVR and the LCVR are
matched).
In an embodiment, the isolated linkage reaction site, e.g., a linkage micro-chamber, does not
include a nucleic acid ng an HCVR or an LCVR from a different cell.
In an embodiment, the isolated linkage reaction site, e.g., a linkage chamber, comprises
a splint oligonucleotide (e.g., a splint oligonucleotide bed herein) that is capable of hybridizing
to a sequence comprising the junction of the HC strand and the LC strand, or a sequence
complementary thereof, to form a duplexed region at the site of ligation.
In an embodiment, the isolated linkage reaction site, e. g., a linkage micro-chamber, ses
a reagent that can covalently link nucleic acids, e.g., a ligase, e.g., a thermostable ligase,
In an aspect, the disclosure features a method of making a nucleic acid sequence comprising a
sequence that encodes an a chain t (AC element) of a TCR 0L chain variable region (ACVR)
and a [3 chain element (BC element) of a TCR [3 chain variable region (BCVR), and wherein the
ACVR and the BCVR are matched, the method comprising:
a) acquiring an isolated production reaction site, e.g., a production micro-chamber,
sing:
i) an a chain (AC) strand, wherein the AC strand is a strand of an or chain double-stranded
cDNA (AC ds cDNA) comprising a segment that s an AC element of the ACVR from a cell,
e.g., an or chain variable region ce (ACVRS); and
ii) a [5 chain (BC) strand, n the BC strand is a strand of a [3 chain ds cDNA (BC ds
cDNA) comprising a segment that s a BC element of the BCVR from the cell, e.g., a [3 chain
variable region sequence (BCVRS), and
b) covalent linking, e. g., ligation, of the first strand to the second strand,
n the isolated production reaction site, e.g., a production micro-chamber, does not
include a nucleic acid encoding an ACVR or a BCVR from a cell other than the cell (e.g., a different
cell, e.g., a different T cell),
thereby making a c acid sequence comprising a sequence that encodes an AC element
of an ACVR and a BC element of a BCVR, wherein the ACVR and the BCVR are matched.
In an embodiment, the AC element comprises, or consists of, an ACVRS, or a functional
fragment thereof (e.g., an antigen binding fragment thereof). In an embodiment, the BC element
comprises, or consists of, a BCVRS, or a functional nt thereof (e.g., an antigen binding
fragment thereof).
In an embodiment, the AC ds cDNA comprises a t that encodes an ACVRS. In an
embodiment, the BC ds cDNA comprises a segment that encodes a BCVRS. In an embodiment, the
AC ds cDNA comprises a segment that encodes an ACVRS, and the BC ds cDNA comprises a
segment that encodes a BCVRS.
In an embodiment, the cell is an immune cell, e.g., a T cell, 6.3., a human T cell. In an
ment, the cell is a mammalian cell or an avian cell.
In an embodiment, the nucleic acid sequence is configured such that, when sed, the AC
element and the BC element (e.g., the ACVRS and the BCVRS) form a functional antigen binding
molecule, e.g., a single chain or a x of a TCR or chain and a [3 chain. In an embodiment, the
antigen binding molecule, e.g., a TCR a chain andior a [3 chain, is functional in vitro, ex vivo, or in
vivo, e.g., as determined by a method or assay described herein.
In an embodiment, acquiring an isolated production reaction site, e.g., a production micro-
chamber, comprises:
a) ing a capture substrate bound to: (i) a first double-stranded cDNA (ds cDNA)
sing a strand that is complementary to a first mRNA that encodes an ACVR from a cell; and
(ii) a second ds cDNA comprising a strand complementary to a second mRNA ng a BCVR
from the cell (the cDNA loaded capture substrate), and
b) maintaining the isolated production reaction site, e.g., the production micro-chamber,
under conditions that allow amplification of the first and second ds cDNAs, to produce: a plurality of
AC ds cDNAs comprising a segment that encodes an AC element of the ACVR from the cell, e. g., an
ACVRS; and a plurality of BC ds cDNAs comprising a segment that encodes a BC element of the
BCVR from the cell, e.g., a BCVRS.
In an embodiment, the AC ds cDNA is identical, or substantially identical, to the first ds
cDNA. For example, the sense strand of the AC ds cDNA is at least 80%, 85 %, 90%, 95%, 98%,
99%, or 100% identical to, or differs by no more than 1, 2, 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50
nucleotides from, the sense strand of the first ds cDNA, and/or the antisense strand of the AC ds
cDNA is at least 80%, 85%, 90%, 95%, 98%, 99%, or 100% identical to, or differs by no more than 1,
2, 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50 tides from, the antisense strand of the first ds cDNA.
In an ment, the BC ds cDNA is identical, or substantially identical, to the second ds
cDNA. For example, the sense strand of the BC ds cDNA is at least 80%, 85%, 90%, 95%, 98%,
99%, or 100% identical to, or differs by no more than 1, 2, 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50
nucleotides from, the sense strand of the second ds cDNA, and/or the antisense strand of the BC ds
cDNA is at least 80%, 85%, 90%, 95%, 98%, 99%, or 100% identical to, or differs by no more than 1,
2, 5, 10, 15, 20, 25, 30, 35, 40, 45 , or 50 nucleotides from, the antisense strand of the second ds
cDNA.
In an embodiment, the AC strand is a sense strand. In an embodiment, the BC strand is a
sense strand. In an embodiment, the AC strand is an antisense strand. In an ment, the BC
strand is an antisense strand. In an embodiment, both the AC strand and the BC strand are sense
strands. In an embodiment, both the AC strand and the BC strand are antisense strands.
In an embodiment, the capture substrate comprises a bead, e.g., a magnetic bead. In an
embodiment, the e substrate comprises a moiety (e.g., an oligonucleotide) which binds to
cDNA, e.g., (i) a moiety which binds to the AC ; (ii) a moiety which binds to the BC ; or
(iii) both (i) and (ii). In an embodiment, the moiety which binds to the AC strand is different from the
moiety which binds to the BC strand, e.g., to facilitate creating conditions favorable to capturing
r levels of each DNA molecule type. In an embodiment, the moiety which binds to the AC
strand is identical to the moiety which binds to the BC strand.
In an embodiment, the first mRNA and the second mRNA are disposed on an mRNA loaded
capture ate.
In an embodiment, the isolated production reaction site, e.g., the production micro-chamber,
comprises: a reagent mixture suitable for producing, from the first and second mRNAs (e.g., after the
first and second mRNAs are released from the loaded mRNA capture substrate into a solution), a first
cDNA comprising a t that encodes an AC element of the ACVR of the cell, e.g., an ACVRS,
and a second cDNA comprising a segment that encodes a BC element of the BCVR of the cell, e.g., a
BCVRS.
In an embodiment, the isolated production reaction site, e.g., production chamber,
comprises primers that mediate the production of the first ds cDNA. In an embodiment, the isolated
production reaction site, e.g., production micro-chamber, comprises primers that mediate the
production of the second ds cDNA.
In an embodiment, a cDNA strand that is mentary to a first mRNA that encodes an
ACVR from a cell is made by reverse transcription of the first mRNA. In an embodiment, a cDNA
strand that is complementary to a second mRNA that encodes a BCVR from a cell is made by reverse
transcription of the second mRNA.
In an ment, the reverse transcription takes place in the isolated production reaction
site, e.g., a production-micro chamber. In an embodiment, the reverse transcription takes place in an
isolated cell reaction site, e.g., a cell ion micro-chamber. In an embodiment, the reverse
transcription takes place outside the isolated production reaction site, e.g., a production micro-
chamber, or outside an isolated cell reaction site, e.g., a cell ion micro-chamber. In an
embodiment, the reverse transcription takes place outside the isolated production reaction site, e. g., a
production-micro chamber, and outside an isolated cell reaction site, e.g., a cell isolation micro-
chamber. In an embodiment, the reverse transcription takes place outside an isolated reaction site,
6.3., outside a micro-chamber.
In an embodiment, the amplification comprises 20 or fewer cycles, e. g., 15 or fewer, 14 or
fewer, 13 or fewer, 12 or fewer, 11 or fewer, 10 or fewer, 9 or fewer, 8 or fewer, 7 or fewer, 6 or
fewer, or 5 or fewer cycles.
In an embodiment, the e transcription and/or amplification uses one or more primers,
e.g., comprising a sequence specific for an ACVRS and/or a BCVRS.
In an embodiment, the reverse transcription and/or ication comprises using two or more
primers that mediate the production of the AC ds cDNA, wherein at least one primer comprises a
nucleotide modification, and n at least one primer does not comprise a nucleotide modification.
In an embodiment, the amplification ses using two or more s that mediate the production
of the BC ds cDNA, wherein at least one primer comprises a nucleotide cation, and wherein at
least one primer does not comprise a nucleotide modification.
In an embodiment, at least one primer ses a nucleotide modification, e.g., which
reduces, e.g., inhibits, DNA synthesis, e.g., by a DNA rase. In an embodiment, at least one
primer does not comprise a nucleotide modification, e.g., which reduces, e.g., inhibits, DNA
synthesis, e.g., by a DNA polymerase.
In an embodiment, the nucleotide ation inhibits a DNA polymerase from extending
the DNA. Without wishing to be bound by theory, it is believed that in an embodiment, any chemical
entity that reduces (e.3., blocks) DNA polymerase extension can be used in accordance with the
methods described herein.
In an embodiment, the nucleotide modification is an insertion of a spacer to the primer, e. g.,
between two adjacent tides in the primer. In an embodiment, the spacer is a flexible . In
an embodiment, the spacer is a carbon spacer (e.g., -(CH2)n-, wherein n=3, 4, 5, 6, 7, 8, 9, 10, or
more), two or more (e.g., three, four, five, six, seven, eight, nine, ten, or more) abasic nucleotides, or a
polyethylene glycol (PEG) spacer. In an embodiment, the spacer is a PEG spacer. In an ment,
the nucleotide modification is 2’-O-methyl, 2’-OH, 2’-NH2, or uracil, e.g., to a ribose.
In an embodiment, the nucleotide modification is located internally or at the 3’ end of the
primer. In an embodiment, at least one primer comprises (i) a first member; (ii) a second member;
and optionally (iii) a third , e.g., comprising a tide modification described herein, e.g.,
located between (i) and (ii).
In an embodiment, the first member is capable of ing with the second member. In an
embodiment, the first member is capable of annealing with the second member in the same primer,
e.g., through intra-molecular hybridization, e.g., to form a n structure comprising a duplex
region of 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, more basepairs. In another
embodiment, the first member is capable of annealing hybridizing with the second member in a
different primer, e. g., through inter-molecular hybridization, e. g., to form a -stranded structure
comprising a duplex region of 4, 5,6, 7, 8, 9, 10, ll, 12, l3, 14, 15, l6, l7, 18, 19, 20, more
basepairs. Without g to be bound by theory, it is believed that in an embodiment, there are at
least two secondary structures that the modified primers can form and tate reduction (e.g.,
prevention) of competition to substrate (e.g., bead) capture. For example, the secondary ure can
be a hairpin-like structure formed by intra-molecular hybridization (within the same ), or the
secondary structure can be a duplex structure formed by inter-molecular hybridization (between two
different primers).
In an embodiment, the first member comprises a sequence that is complementary to the
sequence of an oligonucleotide attached to the e substrate. In an embodiment, the second
member comprises (6.3., from 5’ to 3’) one, two, or all of: (i) a sequence that is complementary to at
least a portion of the first member; (ii) a universal priming sequence (e.g., for PCR ication or
next-generation sequencing); and (iii) a sequence complementary to a target ce, e.g., an
ACVRS and/or a BCVRS. In an embodiment, the universal priming sequence is identical, or
substantially identical, to the ce that is complementary to at least a portion of the first member.
In another embodiment, the universal priming sequence is different from the sequence that is
complementary to at least a portion of the first member. In an embodiment, the second member
comprises a sequence for homologous recombination (e.g., in a yeast or mammalian cell).
In an embodiment, at least one primer comprises a sequence ng at least a portion of a
linker sequence, or a complementary sequence thereof. In an ment, the primer that comprises
a sequence encoding at least a n of a linker sequence, or a mentary sequence thereof, is
phosphorylated, e.g., 5’ phosphorylated. Without wishing to be bound by theory, it is believed that in
an embodiment, any sequence with the general ties of flexibility (e.g., facilitated by glycine)
and hydrophilicity can work effectively in accordance with the methods described herein. Exemplary
linkers can generally have overrepresentation of one or more of Gly, Ser, Thr, or Ala and
underrepresentation of hydrophobic residues, e.g., one or more of Trp, Tyr, Phe, Cys, Met, Leu, or Ile.
The length of the primer may vary, e.g., 3-50 amino acid residues (e.g., 5-45, 10-40, 15-35, 20-30, 10-
20, 10-30, 20-40, or 30-40 amino acid residues). In an embodiment, the linker sequence comprises, or
ts of, ((Gly)m-Ser))n, where m=3, 4, 5, or more and n=1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more. In an
embodiment, the linker sequence comprises, or ts of, (Gly-Gly-Gly-Gly-Ser)n, where n=1, 2, 3,
4, 5, 6, 7, 8, 9, 10, or more.
In an embodiment, the primer is a primer described herein, e. g., in Examples.
In an embodiment, the reverse transcription, the amplification, or both, occurs in a solution in
the isolated production reaction site, e.g., production micro-chamber. In an embodiment, the reverse
transcription, the amplification, or both, does not occur on the substrate (e.g., bead). For example, the
reverse transcription, the amplification, or both, can occur on in a solution within a droplet.
In an embodiment, the AC ds cDNA comprises a 5’ overhang, e. g., a 5’ overhang that is
e of hybridizing to an oligonucleotide attached to a e substrate. In an embodiment, the
AC ds cDNA comprises a blunt end, e.g., a blunt end comprising a 5’ phosphate. In an embodiment,
the BC ds cDNA comprises a 5’ overhang, e.g., a 5’ overhang that is capable of hybridizing to an
oligonucleotide attached to a e substrate. In an embodiment, the BC ds cDNA comprises a
blunt end, e.g., a blunt end comprising a 5’ phosphate. In an embodiment, the AC ds cDNA and the
BC ds cDNA comprise sticky ends, e.g., both have 5’ overhangs.
In an embodiment, the AC strand and the BC strand are covalently linked, e.g., ligated, to
produce a single stranded nucleic acid sequence, wherein the AC and BC strands are both sense
strands or both antisense strands. In an embodiment, a denatured AC strand of the AC ds cDNA to a
red BC strand of the BC ds cDNA are covalently , e.g., ligated, wherein the AC and BC
strands are both sense strands or both antisense strands. In an ment, the AC strand is present
in the AC ds cDNA and the BC strand is present in the BC ds cDNA, and wherein the AC ds cDNA
and the BC ds cDNA are covalently linked, e.g., ligated, e.g., to produce a double stranded nucleic
acid ce.
In an embodiment, the covalent linking, e.g., ligation, occurs in the isolated production
reaction site. In an embodiment, the isolated production reaction site, e.g., a production micro-
chamber, or the isolated linkage reaction site, e.g., a linkage micro-chamber, comprises a reagent that
is e of covalently linking, e.g., ligating, the AC and BC strands or the AC and BC ds cDNAs.
In an embodiment, the isolated production reaction site, e.g., a production micro-chamber comprises
an enzyme that covalently couples the AC and BC strands or the AC and BC ds cDNAs. In an
embodiment, the enzyme is a ligase, e.g., a l stable ligase. In an embodiment, the covalent
linking comprises ligase thermocycling.
In an embodiment, the covalent linking, e. g., ligation, occurs in a site different from the
isolated production reaction site, e.g., occurs in an isolated linkage reaction site, e.g., a linkage micro-
r. In an embodiment, the AC strand and the BC strand are transferred from the isolated
production site to the isolated linkage reaction site, e.g., a linkage micro-chamber, and the covalent
linking occurs in the ed linkage reaction site, e.g., a linkage micro-chamber. In an embodiment,
the isolated e reaction site, e.g., a linkage micro-chamber, comprises a reagent that is capable of
covalently linking, e.g., ligating, the AC and BC strands or the AC and BC ds cDNAs. In an
embodiment, the isolated linkage on site, e.g., a linkage micro-chamber, comprises an enzyme
that covalently couples the AC and BC strands or the AC and BC ds cDNAs. In an ment, the
enzyme is a ligase, e.g., a thermal stable ligase. In an embodiment, the covalent linking comprises
ligase thermocycling.
In an embodiment, the covalent linking, e.g., ligation, comprises: (a) g the isolated
linkage reaction site, e. g., the linkage chamber, under conditions (e.g., at 950C) that allow
denaturation of the AC strand and the BC strand; (b) cooling the isolated linkage reaction site, e. g.,
the e micro-chamber, under conditions (e.g., at C) that allow hybridization of the splint
oligonucleotide to the AC strand and the BC strand; (c) maintaining the isolated linkage reaction site,
6.3., the linkage micro-chamber, under conditions (e.g., at 45-650C) that allow ligation of the AC
strand and the BC strand (e.g., formation of phosphodiester bond between the AC strand and the BC
strand); and (d) repeating steps (a), (b), and (c) sequentially for 2, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50,
or more cycles.
In an ment, the AC strand and the BC strand are covalently linked, e.g., ligated, in the
ce of a splint oligonucleotide. In an embodiment, the splint oligonucleotide is hybridized to a
sequence comprising the junction of the AC strand and the BC strand, or a sequence complementary
thereof, and forms a duplexed region at the site of on. In an embodiment, the splint
oligonucleotide comprises a modification (e.g., an NHZ group) that inhibits DNA synthesis, e.g., by a
DNA polymerase. In an embodiment, the modification is at the 3’ end of the splint oligonucleotide.
In an embodiment, a strand complimentary to the covalently linked, e.g., d, AC and BC
strands is produced by amplification.
In an embodiment, the method, e.g., the step of covalent e, does not include a step of
overlap extension polymerase chain reaction (OE-PCR), also known as splicing by overlap ion
or splicing by overhang extension (SOE) PCR.
In an embodiment, the method further comprises, prior to acquiring the ed production
reaction site, e.g., a production micro-chamber, acquiring an mRNA loaded capture substrate.
In an ment, acquiring the mRNA loaded capture substrate comprising: a) acquiring an
isolated cell on site, e.g., a cell isolation chamber, comprising: i) a cell; and ii) a capture
substrate capable of binding a first mRNA ng an ACVR from the cell and a second mRNA
encoding a BCVR from the cell; and b) ining the isolated cell reaction site, e.g., the cell
isolation micro-chamber, under conditions that allow lysis of the cell and binding of the capture
substrate with the first mRNA and the second mRNA to form the mRNA loaded capture substrate,
n the isolated cell reaction site, e.g., cell isolation micro-chamber, does not include a nucleic
acid encoding an ACVR or a BCVR from a cell other than the cell (e.g., a ent cell).
In an embodiment, the isolated cell reaction site, e.g., cell isolation micro-chamber, comprises
a lysing reagent, e.g., a detergent. In an embodiment, the cell is lysed by heat or an . In an
embodiment, the capture substrate comprises a moiety (e.g., an oligonucleotide) which binds mRNA,
e.g., an oligo(dT).
In an ment, the method r comprises releasing the mRNA loaded capture
substrate from the isolated cell reaction site, e.g., the cell isolation micro-chamber. In an
embodiment, the releasing step is performed in the presence of a poly(dA) or poly(dT)
oligonucleotide, e.g., to reduce cross-binding of non-captured mRNA.
In an embodiment, the mRNA loaded capture substrate is transferred from the isolated cell
reaction site, e.g., the cell isolation micro-chamber, to the isolated production reaction site, e.g., the
production micro-chamber.
In an ment, the method further comprises releasing the nucleic acid ce from the
isolated production reaction site, e.g., the production micro-chamber. In an embodiment, the method
further comprises amplifying the nucleic acid sequence. In an embodiment, amplification of the
nucleic acid sequence occurs outside the isolated production reaction site, e.g., the production micro-
chamber, e. g., after the nucleic acid is ed from the isolated production reaction site, e.g., the
tion micro-chamber. In an embodiment, amplification of the nucleic acid sequence occurs at
the isolated production reaction site, e.g., the production micro-chamber.
In an embodiment, the method further ses sequencing all or a portion of the nucleic
acid sequence.
In an embodiment, the method further comprises inserting all or a portion of nucleic acid
sequence into a vector. In an embodiment, the vector supplies an additional AC t or BC
element not included in the nucleic acid sequence. In an embodiment, the method further comprises
expressing the vector.
In an embodiment, the method further comprises expressing the nucleic acid ce to
produce a polypeptide comprising a segment that encodes an AC element of the ACVR, e. g., an
ACVRS, and a segment that encodes a BC element of the BCVR, e.g., a BCVRS. In an embodiment,
the BC element is N—terminal to the AC element in the polypeptide. In an embodiment, the AC
element is C-terminal to the BC element in the polypeptide.
In an embodiment, the method further comprises contacting the polypeptide with an n.
In an embodiment, the method r comprises determining if the polypeptide binds the antigen, in
vitro, ex vivo, or in viva, e.g., by a method or assay described herein.
In an embodiment, the disclosure features a method of making a nucleic acid sequence
sing a sequence that encodes a TCR 0L chain element (AC element) of TCR 0L chain le
region (ACVR) and a TCR [3 chain element (BC element) of a TCR [3 chain le region (BCVR),
and wherein the ACVR and BCVR are matched, comprising:
a) acquiring an isolated cell on site (e.g., an isolated cell reaction site described herein),
e.g., a cell isolation micro-chamber, sing: i) a cell (e.g., a cell described herein); and ii) a
capture substrate (e.g., a capture ate bed herein) capable of binding a first mRNA
encoding an ACVR from the cell and a second mRNA encoding a BCVR from the cell;
b) maintaining the isolated cell reaction site, e.g., the cell isolation micro-chamber, under
conditions that allow lysis of the cell and binding of the capture substrate with the first mRNA and the
second mRNA to form an mRNA loaded capture substrate,
wherein the isolated cell reaction site, e.g., cell isolation micro-chamber, does not include a
nucleic acid encoding an ACVR or a BCVR from a cell other than the cell (e.g., a different cell);
c) contacting the mRNA loaded capture substrate with a reaction mixture, e.g., a reaction
mixture sing reverse transcriptase, that uses the loaded mRNA as a template to make cDNA
(this can occur, e.g., in the isolated cell reaction site, in an isolated production reaction site, or in
neither, e.g., not in an isolated reaction site);
d) acquiring an isolated production reaction site (e. g., an isolated tion reaction site
described herein), e.g., a production micro-chamber, sing: i) a TCR 0L chain (AC) ,
wherein the AC strand is a strand of a TCR a chain double-stranded cDNA (AC ds cDNA)
comprising a segment that encodes an AC element of the ACVR from the cell, e.g., a TCR 0L chain
variable region sequence (ACVRS); and ii) a TCR [5 chain (BC) strand, n the BC strand is a
strand of a TCR [3 chain double-stranded cDNA (BC ds cDNA) comprising a segment that encodes a
BC element of the BCVR from the cell, e. g., a TCR [3 chain variable region sequence (BCVRS),
wherein the isolated production reaction site, e.g., a production micro-chamber, does not
include a nucleic acid encoding an ACVR or a BCVR from a cell other than the cell (e.g., a different
cell); and
e) covalent linking, e.g., on, of the AC strand to the BC strand.
In an embodiment, one or more (e.g., two, three, four, or all) of the steps a)-e) are performed
in accordance with a method described herein. In an embodiment, each of the steps a)-e) is performed
in accordance with a method described herein.
In an aspect, the disclosure features a method of making a nucleic acid ce comprising a
ce that encodes a TCR 0c chain element (AC element) of a TCR a chain variable region
(ACVR) and a TCR B chain element (BC element) of a TCR [3 chain variable region (BCVR), and
wherein the ACVR and BCVR are matched, comprising:
a) acquiring an isolated cell reaction site (e.g., an isolated cell reaction site described herein),
e.g., a cell isolation micro-chamber, sing: i) a cell (e.g., a cell described herein); and ii) a
capture substrate (e. g., a capture substrate bed herein) capable of binding a first mRNA
encoding an ACVR from the cell and a second mRNA encoding a BCVR from the cell;
b) ining the isolated cell reaction site, e.g., the cell isolation micro-chamber, under
conditions that allow lysis of the cell and binding of the capture substrate with the first mRNA and the
second mRNA to form the mRNA loaded capture substrate,
wherein the isolated cell on site, e.g., cell isolation micro-chamber, does not include a
nucleic acid encoding an ACVR or a BCVR from a cell other than the cell (e.g., a ent cell);
c) acquiring an isolated production reaction site (e.g., an isolated production reaction site
described herein), e.g., a production micro-chamber, comprises: contacting the mRNA loaded capture
substrate with a reaction mixture, e.g., a on mixture comprising e transcriptase, that uses
the loaded mRNA as a template, to produce: a first double-stranded cDNA (ds cDNA) comprising a
strand that is complementary to a first mRNA that encodes an ACVR from a cell; and a second ds
cDNA comprising a strand complementary to a second mRNA encoding a BCVR from the cell (the
cDNA loaded capture substrate);
wherein the isolated production reaction site, e.g., a tion micro-chamber, does not
include a nucleic acid encoding an ACVR or a BCVR from a cell other than the cell (e. g., a different
cell).
d) maintaining the isolated production reaction site, e.g., the production micro-chamber,
under conditions that allow amplification of the first and second ds cDNAs, to produce: a plurality of
AC ds cDNAs comprising a segment that encodes an AC element of the ACVR from the cell, e.g., an
ACVRS; and a plurality of BC ds cDNAs comprising a segment that encodes a BC element of the
BCVR from the cell, e.g., a BCVRS;
e) acquiring an isolated linkage reaction site (e.g., an isolated linkage on site bed
herein), e.g., a linkage micro-chamber, comprising: covalent linking, e. g., ligation, of a strand of the
AC ds cDNA (AC strand) to a strand of the BC ds cDNA (BC strand), n the AC and BC
strands are both sense strands or nse strands; and
f) amplifying the covalently linked, e.g., ligated, AC and BC s.
In an embodiment, one or more (e. g., two, three, four, five, or all) of the steps a)-f) are
performed in accordance with a method described herein. In an embodiment, each of the steps a)-f) is
performed in ance with a method described herein.
In an aspect, the disclosure es a method of making a library comprising a plurality of
unique members, the method sing:
making the plurality of members, wherein each of the members comprises a sequence that
encodes a TCR a chain t (AC element) of a TCR a chain variable region (ACVR) and a TCR [3
chain element (BC element) of a TCR [3 chain variable region (BCVR), and wherein the ACVR and
BCVR are matched, made by a method described ,
wherein each unique nucleic acid sequence of the plurality comprises an AC element and a
BC element from a different unique cell (e. g., a cell described herein),
thereby making a library comprising a plurality of unique members.
In an embodiment, the plurality of unique members comprises at least 104, 105, 106, 107, 108,
or 109 unique s. In an embodiment, the ity of unique members comprises 104 to 109, 104
to 108, 104 to 107, 104 to 106, 104 to 105, 108 to 109, 107 to 109, 106 to 109, 105 to 109, 105 to 10", 106 to
107, 104 to 105, 105 to 106, 106 to 107, 107 to 108, or 108 to 109 unique members. In an embodiment, at
least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%, of the members in the library are unique
s (which encode matched AC t and BC element sequences). In an embodiment, less
than 20%, 15%, 10%, 5%, 4%, 3%, 2%, or 1%, of the members in the y are unique members
(which encode matched AC element and BC element sequences).
In an aspect, the disclosure features a library comprising a plurality of unique members,
wherein,
i) each unique member of the plurality comprises a segment that encodes an AC element, e.g.,
an ACVRS, and a segment that encodes a BC element, e.g., a BCVRS, wherein the AC element and
the BC t in each unique member is matched;
ii) each unique member of the plurality comprises a segment that s an AC element,
e.g., an ACVRS, and a segment that encodes a BC element, e.g., a BCVRS, from a different unique
cell; and
iii) the library comprises one or more (e.g., two, three, four, or all) of the following
properties:
a) the library is made by a method described herein;
b) the plurality of unique members comprises at least 104, 105, 106, 107, 108, or 109 unique
nucleic acid sequences;
c) the plurality of unique members comprises 104 to 109, 104 to 108, 104 to 107, 104 to 106, 104
to 105, 108 to 109, 107 to 109, 106 to 109, 105 to 109, 105 to 103, 106 to 107, 104 to 105, 105 to 106, 106 to
107, 107 to 108, or 108 to 109 unique members;
d) at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%, of the members in the
library are unique members (which encode matched AC element and BC element sequences); or
WO 19402
e) less than 20%, 15%, 10%, 5%, 4%, 3%, 2%, or 1%, of the members in the library are
unique members (which encode matched AC element and BC element sequences).
In an embodiment, each unique member of the plurality is configured such that, when
expressed, the AC element, e.g., the ACVRS, and the BC element, e.g., the BCVRS, form a functional
antigen g molecule, e.g., a single chain or a complex of a TCR a chain and a [3 chain.
In an embodiment, the library is a display library. In an embodiment, each of the members of
the plurality further encodes a polypeptide that results in display of the member on the surface of a
display entity. In an embodiment, the library is a phage display library. In an embodiment, the
library is a yeast display library. In an embodiment, the library is a mammalian display library.
In an aspect, the disclosure features a method of making a binding polypeptide (e.g., a
polypeptide comprising an AC element and a BC element), the method comprising: a) acquiring a
library described herein, e.g., by a method described herein; and b) expressing a polypeptide encoded
by a unique nucleic acid of the library.
In an embodiment, the method further comprises contacting the polypeptide with an antigen.
In an embodiment, the method r comprises retrieving (e.g., isolating or ing) the nucleic
acid that encodes a polypeptide that binds the antigen.
In an aspect, the disclosure features an isolated tion reaction site, e.g., a production
micro-chamber, which is an isolated production reaction site described herein (e.g., comprising a
nucleic acid encoding an ACVR and a nucleic acid encoding a BCVR, n the ACVR and the
BCVR are matched).
In an embodiment, the isolated production on site, e.g., a production micro-chamber,
does not e a c acid encoding an ACVR or a BCVR from a different cell.
In an embodiment, the isolated tion reaction site, e.g., a production micro-chamber,
comprises one, two, or all of: (i) one or more primers ic to V gene ces of the AC and BC;
(ii) one or more primers c to overhangs introduced onto the AC and BC cDNAs; or (iii) one or
more s sing a first member, a second member, and a third member sing a
nucleotide modification (e.g., a spacer) located between the first and second members, wherein the
first member is capable of annealing with the second member of the same primer or a different
primer, e.g., forming a structure sing a duplex region of 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15,
16, 17, 18, 19, 20, more basepairs.
In an embodiment, the isolated production reaction site, e.g., a production micro-chamber,
does not comprise a reagent that can covalently link nucleic acids, e.g., a ligase, e.g., a thermostable
1i gase. In another embodiment, the isolated production reaction site, e. g., a production micro-
chamber, comprises a reagent that can covalently link nucleic acids, e.g., a ligase, e.g., a thermostable
ligase.
In an , the disclosure features a self-annealing oligonucleotide comprising a first
member, a second , and third member comprising a nucleotide ation (e. g., a spacer)
located between the first and second members, wherein the first member is e of ing with
the second member of the same oligonucleotide (e.g., for a method of making a nucleic acid sequence
comprising a sequence that s an AC element of an ACVR and a BC t of a BCVR,
n the ACVR and BCVR are matched).
In an embodiment, the first and second members are capable of forming a hairpin structure
sing a duplex region of 4, 5,6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, more
basepairs. In an embodiment, the first member is 5-40 nucleotides, e.g., 5-10, 5-20, 5-30, 30-40, 20-
40, 10-30, 10-30, or 15-25 nucleotides, in length. In an embodiment, the second member is 5-40
nucleotides, e.g., 5-10, 5-20, 5-30, 30-40, 20-40, 10-30, 10-30, or 15-25 nucleotides, in length.
In an embodiment, the spacer is a spacer described , e.g., a flexible spacer or a PEG
spacer.
In an embodiment, the first member ses a sequence that is complementary to the
sequence of an oligonucleotide attached to a capture substrate.
In an embodiment, the second member comprises (e.g., from 5’ to 3’) one, two, or all of: (i) a
sequence that is complementary to at least a portion of the first ; (ii) a universal priming
sequence (e.g., for PCR amplification or next-generation sequencing); and (iii) a sequence
complementary to a target sequence, e.g., an ACVRS and/or a BCVRS. In an embodiment, the
universal priming sequence is identical, or substantially identical, to the sequence that is
complementary to at least a n of the first member. In another embodiment, the universal
priming sequence is different from the sequence that is complementary to at least a portion of the first
member. In an embodiment, the second member comprises a sequence for homologous
ination (e.g., in a yeast or mammalian cell).
In an aspect, the disclosure features an isolated linkage reaction site, e.g., a linkage micro-
chamber, which is an isolated linkage reaction site described herein (e.g., comprising a nucleic acid
encoding an ACVR and a nucleic acid ng a BCVR, wherein the ACVR and the BCVR are
matched).
In an embodiment, the isolated linkage reaction site, e.g., a linkage micro-chamber, does not
include a nucleic acid encoding an ACVR or a BCVR from a different cell.
In an embodiment, the isolated linkage reaction site, 6. g., a linkage micro-chamber, comprises
a splint oligonucleotide (e.g., a splint oligonucleotide described herein) that is capable of hybridizing
to a sequence comprising the junction of the AC strand and the BC strand, or a sequence
complementary thereof, to form a duplexed region at the site of ligation.
In an embodiment, the isolated linkage reaction site, e.g., a linkage micro-chamber, comprises
a reagent that can covalently link nucleic acids, e.g., a ligase, e.g., a stable ligase.
In an aspect, the disclosure es a method of making a nucleic acid sequence comprising a
sequence that encodes a 7 chain element (GC element) of a TCR V chain variable region (GCVR) and
a 5 chain element (DC element) of a TCR 5 chain variable region (DCVR), and wherein the GCVR
and the DCVR are matched, the method sing:
a) acquiring an isolated production reaction site, e.g., a production micro-chamber,
comprising:
i) an y chain (GC) strand, wherein the GC strand is a strand of an 7 chain -stranded
cDNA (GC ds cDNA) comprising a segment that encodes a GC element of the GCVR from a cell,
e.g., an 7 chain variable region sequence (GCVRS); and
ii) a 5 chain (DC) , wherein the DC strand is a strand of a 5 chain ds cDNA (DC ds
cDNA) comprising a segment that encodes a DC element of the DCVR from the cell, e. g., a 5 chain
variable region sequence (DCVRS), and
b) covalent g, e.g., ligation, of the first strand to the second strand,
wherein the isolated production reaction site, e.g., a production micro-chamber, does not
include a nucleic acid encoding a GCVR or a DCVR from a cell other than the cell (e.g., a different
cell, e.g., a different T cell),
thereby making a c acid sequence comprising a sequence that encodes a GC element of
a GCVR and a DC element of a DCVR, wherein the GCVR and the DCVR are matched.
In an embodiment, the GC element comprises, or consists of, a GCVRS, or a functional
fragment thereof (e.g., an antigen binding fragment thereof). In an embodiment, the DC element
ses, or consists of, a DCVRS, or a functional fragment thereof (e.g., an antigen binding
fragment thereof).
In an ment, the GC ds cDNA comprises a segment that encodes a GCVRS. In an
embodiment, the DC ds cDNA ses a segment that encodes a DCVRS. In an embodiment, the
GC ds cDNA comprises a segment that encodes a GCVRS, and the DC ds cDNA comprises a
segment that encodes a DCVRS.
In an embodiment, the cell is an immune cell, e.g., a T cell, 6.3., a human T cell. In an
ment, the cell is a mammalian cell or an avian cell.
In an embodiment, the nucleic acid sequence is configured such that, when expressed, the GC
element and the DC element (e.g., the GCVRS and the DCVRS) form a functional antigen binding
molecule, e.g., a single chain or a complex of a TCR 7 chain and a 5 chain. In an embodiment, the
n binding molecule, e.g., a TCR V chain and/or a 5 chain, is functional in vitro, ex vivo, or in
viva, e.g., as determined by a method or assay described herein.
In an ment, acquiring an isolated production reaction site, e.g., a production micro-
chamber, comprises:
a) acquiring a e substrate bound to: (i) a first double-stranded cDNA (ds cDNA)
comprising a strand that is complementary to a first mRNA that encodes a GCVR from a cell; and (ii)
a second ds cDNA comprising a strand complementary to a second mRNA encoding a DCVR from
the cell (the cDNA loaded capture substrate), and
b) maintaining the isolated production reaction site, e.g., the production chamber,
under conditions that allow cation of the first and second ds cDNAs, to e: a plurality of
GC ds cDNAs comprising a segment that encodes a GC element of the GCVR from the cell, e.g., a
GCVRS; and a plurality of DC ds cDNAs comprising a segment that s a DC element of the
DCVR from the cell, e.g., a DCVRS.
In an embodiment, the GC ds cDNA is identical, or ntially identical, to the first ds
cDNA. For example, the sense strand of the GC ds cDNA is at least 80%, 85 %, 90%, 95%, 98%,
99%, or 100% identical to, or differs by no more than 1, 2, 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50
nucleotides from, the sense strand of the first ds cDNA, and/or the antisense strand of the GC ds
cDNA is at least 80%, 85%, 90%, 95%, 98%, 99%, or 100% identical to, or differs by no more than 1,
2, 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50 nucleotides from, the antisense strand of the first ds cDNA.
In an embodiment, the DC ds cDNA is identical, or substantially identical, to the second ds
cDNA. For example, the sense strand of the DC ds cDNA is at least 80%, 85 %, 90%, 95%, 98%,
99%, or 100% identical to, or differs by no more than 1, 2, 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50
nucleotides from, the sense strand of the second ds cDNA, and/or the antisense strand of the DC ds
cDNA is at least 80%, 85%, 90%, 95%, 98%, 99%, or 100% identical to, or s by no more than 1,
2, 5, 10, 15, 20, 25, 30, 35, 40, 45 , or 50 nucleotides from, the antisense strand of the second ds
cDNA.
In an embodiment, the GC strand is a sense strand. In an embodiment, the DC strand is a
sense strand. In an embodiment, the GC strand is an antisense strand. In an embodiment, the DC
strand is an antisense strand. In an embodiment, both the GC strand and the DC strand are sense
strands. In an embodiment, both the GC strand and the DC strand are antisense strands.
In an embodiment, the capture substrate comprises a bead, e.g., a magnetic bead. In an
embodiment, the capture substrate ses a moiety (e.g., an oligonucleotide) which binds to
cDNA, e.g., (i) a moiety which binds to the GC strand; (ii) a moiety which binds to the DC strand; or
(iii) both (i) and (ii). In an embodiment, the moiety which binds to the GC strand is different from the
moiety which binds to the DC strand, e.g., to facilitate creating conditions favorable to ing
similar levels of each DNA molecule type. In an embodiment, the moiety which binds to the GC
strand is identical to the moiety which binds to the DC strand.
In an embodiment, the first mRNA and the second mRNA are ed on an mRNA loaded
capture substrate.
In an embodiment, the isolated production reaction site, e. g., the production micro-chamber,
comprises: a reagent mixture suitable for producing, from the first and second mRNAs (e.g., after the
first and second mRNAs are released from the mRNA loaded capture substrate into a solution), a first
cDNA comprising a segment that encodes a GC element of the GCVR of the cell, e.g., a GCVRS, and
a second cDNA comprising a t that encodes a DC element of the DCVR of the cell, e.g., a
DCVRS.
In an embodiment, the isolated production reaction site, e.g., production micro-chamber,
ses primers that mediate the production of the first ds cDNA. In an embodiment, the isolated
production reaction site, e.g., production micro-chamber, comprises primers that mediate the
production of the second ds cDNA.
In an embodiment, a cDNA strand that is complementary to a first mRNA that encodes a
GCVR from a cell is made by reverse transcription of the first mRNA. In an embodiment, a cDNA
strand that is complementary to a second mRNA that encodes a DCVR from a cell is made by reverse
transcription of the second mRNA.
In an embodiment, the reverse ription takes place in the isolated production on
site, e.g., a production-micro chamber. In an embodiment, the e transcription takes place in an
isolated cell reaction site, e.g., a cell isolation micro-chamber. In an embodiment, the reverse
transcription takes place outside the isolated production reaction site, e.g., a production micro-
chamber, or outside an isolated cell reaction site, e.g., a cell isolation micro-chamber. In an
embodiment, the reverse transcription takes place outside the isolated production reaction site, e.g., a
production-micro chamber, and e an isolated cell reaction site, e.g., a cell isolation micro-
chamber. In an ment, the reverse transcription takes place outside an isolated reaction site,
e.g., e a micro-chamber.
In an embodiment, the amplification comprises 20 or fewer cycles, e.g., 15 or fewer, 14 or
fewer, 13 or fewer, 12 or fewer, 11 or fewer, 10 or fewer, 9 or fewer, 8 or fewer, 7 or fewer, 6 or
fewer, or 5 or fewer cycles.
In an embodiment, the reverse transcription and/or amplification uses one or more primers,
e.g., comprising a sequence specific for a GCVRS and/or a DCVRS.
In an embodiment, the reverse transcription and/or amplification comprises using two or more
primers that mediate the tion of the GC ds cDNA, wherein at least one primer comprises a
nucleotide modification, and wherein at least one primer does not comprise a nucleotide modification.
In an embodiment, the cation comprises using two or more primers that mediate the tion
of the DC ds cDNA, wherein at least one primer comprises a nucleotide modification, and wherein at
least one primer does not comprise a nucleotide modification.
In an ment, at least one primer comprises a nucleotide modification, e. g., which
reduces, e.g., inhibits, DNA synthesis, e.g., by a DNA polymerase. In an embodiment, at least one
primer does not comprise a tide modification, e.g., which reduces, e.g., inhibits, DNA
synthesis, e.g., by a DNA polymerase.
In an embodiment, the nucleotide ation inhibits a DNA rase from extending
the DNA. Without wishing to be bound by theory, it is believed that in an embodiment, any chemical
entity that reduces (e.g., blocks) DNA rase extension can be used in accordance with the
methods described herein.
In an ment, the tide modification is an insertion of a spacer to the , e.g.,
between two adjacent nucleotides in the primer. In an embodiment, the spacer is a flexible spacer. In
an embodiment, the spacer is a carbon spacer (e.g., -(CH2)n-, wherein n=3, 4, 5, 6, 7, 8, 9, 10, or
more), two or more (e.g., three, four, five, six, seven, eight, nine, ten, or more) abasic nucleotides, or a
polyethylene glycol (PEG) . In an embodiment, the spacer is a PEG spacer. In an ment,
the nucleotide modification is 2’-O-methyl, 2’-OH, 2’-NH2, or uracil, e.g., to a ribose.
In an embodiment, the nucleotide modification is located internally or at the 3’ end of the
primer. In an embodiment, at least one primer comprises (i) a first member; (ii) a second member;
and optionally (iii) a third member, e.g., comprising a nucleotide modification described herein, e.g.,
located n (i) and (ii).
In an embodiment, the first member is capable of annealing with the second member. In an
embodiment, the first member is capable of annealing with the second member in the same primer,
e.g., through intra-molecular hybridization, e.g., to form a hairpin structure comprising a duplex
region of 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, more basepairs. In another
embodiment, the first member is capable of annealing hybridizing with the second member in a
different primer, e.g., through inter-molecular hybridization, e.g., to form a double-stranded structure
comprising a duplex region of 4, 5,6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, more
basepairs. Without wishing to be bound by theory, it is believed that in an embodiment, there are at
least two ary structures that the modified primers can form and facilitate reduction (e.g.,
prevention) of competition to substrate (e.g., bead) capture. For e, the secondary structure can
be a hairpin-like ure formed by intra-molecular hybridization (within the same primer), or the
secondary structure can be a duplex structure formed by inter-molecular hybridization (between two
different primers).
In an ment, the first member ses a sequence that is complementary to the
sequence of an oligonucleotide attached to the capture substrate. In an embodiment, the second
member comprises (e.g., from 5’ to 3’) one, two, or all of: (i) a sequence that is complementary to at
least a portion of the first member; (ii) a universal priming sequence (e.g., for PCR amplification or
eneration sequencing); and (iii) a sequence complementary to a target sequence, e.g., a GCVRS
andior a DCVRS. In an embodiment, the universal priming sequence is identical, or substantially
identical, to the sequence that is complementary to at least a portion of the first member. In another
embodiment, the universal priming sequence is different from the sequence that is complementary to
at least a portion of the first member. In an embodiment, the second member ses a sequence
for homologous ination (e.g., in a yeast or mammalian cell).
In an embodiment, at least one primer comprises a sequence encoding at least a portion of a
linker sequence, or a complementary sequence thereof. In an embodiment, the primer that comprises
a sequence encoding at least a portion of a linker ce, or a complementary sequence thereof, is
phosphorylated, e.g., 5’ phosphorylated. Without wishing to be bound by theory, it is believed that in
an embodiment, any ce with the general properties of flexibility (e.g., facilitated by glycine)
and hydrophilicity can work effectively in accordance with the methods described herein. Exemplary
linkers can generally have overrepresentation of one or more of Gly, Ser, Thr, or Ala and
underrepresentation of hydrophobic es, e.g., one or more of Trp, Tyr, Phe, Cys, Met, Leu, or 116.
The length of the primer may vary, e.g., 3-50 amino acid residues (e.g., 5-45, 10-40, 15-35, 20-30, 10-
, 10-30, 20-40, or 30-40 amino acid residues). In an ment, the linker sequence comprises, or
consists of, ((Gly)m-Ser))n, where m=3, 4, 5, or more and n=1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more. In an
embodiment, the linker sequence comprises, or consists of, (Gly-Gly-Gly-Gly-Ser)n, where n=1, 2, 3,
4, 5, 6, 7, 8, 9, 10, or more.
In an embodiment, the primer is a primer described herein, e.g., in Examples.
In an embodiment, the reverse transcription, the amplification, or both, occurs in a on in
the isolated production reaction site, e.g., tion micro-chamber. In an embodiment, the reverse
transcription, the amplification, or both, does not occur on the substrate (e.g., bead). For example, the
reverse transcription, the amplification, or both, can occur on in a solution within a droplet.
In an embodiment, the GC ds cDNA comprises a 5’ overhang, e.g., a 5’ overhang that is
capable of hybridizing to an oligonucleotide attached to a capture substrate. In an embodiment, the
GC ds cDNA comprises a blunt end, e.g., a blunt end comprising a 5’ phosphate. In an embodiment,
the DC ds cDNA ses a 5’ overhang, e.g., a 5’ overhang that is capable of hybridizing to an
ucleotide attached to a capture ate. In an embodiment, the DC ds cDNA comprises a
blunt end, e. g., a blunt end comprising a 5’ phosphate. In an embodiment, the GC ds cDNA and the
DC ds cDNA comprise sticky ends, e.g., both have 5’ overhangs.
In an embodiment, the GC strand and the DC strand are covalently linked, e.g., ligated, to
produce a single stranded nucleic acid sequence, wherein the GC and DC strands are both sense
strands or both antisense strands. In an embodiment, a red GC strand of the GC ds cDNA to a
denatured DC strand of the DC ds cDNA are covalently , e.g., ligated, wherein the GC and DC
strands are both sense strands or both nse strands. In an embodiment, the GC strand is present
in the GC ds cDNA and the DC strand is present in the DC ds cDNA, and wherein the GC ds cDNA
and the DC ds cDNA are covalently linked, e.g., ligated, e.g., to produce a double stranded nucleic
acid ce.
In an embodiment, the covalent linking, e.g., ligation, occurs in the isolated production
reaction site. In an embodiment, the isolated tion reaction site, e.g., a production micro-
r, or the isolated linkage reaction site, e.g., a linkage micro-chamber, comprises a reagent that
is capable of covalently linking, e.g., ligating, the GC and DC strands or the GC and DC ds cDNAs.
In an ment, the ed production reaction site, e.g., a production micro-chamber comprises
an enzyme that covalently couples the GC and DC s or the GC and DC ds cDNAs. In an
embodiment, the enzyme is a ligase, e. g., a thermal stable ligase. In an embodiment, the covalent
g comprises ligase thermocycling.
In an embodiment, the covalent linking, e. 3., ligation, occurs in a site different from the
isolated production reaction site, e.g., occurs in an isolated linkage reaction site, e.g., a linkage micro-
chamber. In an embodiment, the GC strand and the DC strand are transferred from the ed
production site to the isolated linkage reaction site, e.g., a linkage micro-chamber, and the nt
linking occurs in the isolated linkage on site, e.g., a linkage micro-chamber. In an embodiment,
the isolated linkage reaction site, e.g., a linkage micro-chamber, comprises a reagent that is capable of
covalently linking, e.g., ligating, the GC and DC strands or the GC and DC ds cDNAs. In an
embodiment, the isolated linkage reaction site, e. g., a linkage micro-chamber, comprises an enzyme
that ntly couples the GC and DC s or the GC and DC ds cDNAs. In an embodiment, the
enzyme is a ligase, e.g., a thermal stable ligase. In an embodiment, the covalent linking comprises
ligase thermocycling.
In an embodiment, the covalent linking, e. 3., ligation, comprises: (a) heating the isolated
linkage reaction site, e. g., the linkage micro-chamber, under conditions (e.g., at 950C) that allow
denaturation of the GC strand and the DC ; (b) cooling the isolated linkage reaction site, e.g.,
the e micro-chamber, under conditions (e.g., at 50-65°C) that allow hybridization of the splint
oligonucleotide to the GC strand and the DC strand; (c) maintaining the isolated linkage reaction site,
e.g., the linkage micro-chamber, under conditions (e.g., at 45-650C) that allow ligation of the GC
strand and the DC strand (e.g., formation of phosphodiester bond n the GC strand and the DC
strand); and (d) repeating steps (a), (b), and (c) sequentially for 2, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50,
or more cycles.
In an embodiment, the GC strand and the DC strand are covalently linked, e.g., ligated, in the
presence of a splint oligonucleotide. In an embodiment, the splint oligonucleotide is hybridized to a
sequence comprising the junction of the GC strand and the DC strand, or a sequence complementary
thereof, and forms a duplexed region at the site of ligation. In an ment, the splint
oligonucleotide comprises a modification (e.g., an NH2 group) that inhibits DNA synthesis, e.g., by a
DNA polymerase. In an embodiment, the ation is at the 3’ end of the splint oligonucleotide.
In an embodiment, a strand complimentary to the ntly linked, e.g., ligated, GC and DC
strands is produced by amplification.
In an embodiment, the method, e.g., the step of covalent linkage, does not include a step of
overlap extension polymerase chain on (OE-PCR), also known as splicing by p extension
or splicing by overhang extension (SOE) PCR.
In an embodiment, the method further comprises, prior to ing the isolated tion
reaction site, e.g., a production micro-chamber, ing an mRNA loaded e substrate.
In an ment, acquiring the mRNA loaded capture substrate comprising: a) acquiring an
isolated cell reaction site, e.g., a cell ion micro-chamber, comprising: i) a cell; and ii) a capture
substrate capable of binding a first mRNA ng a GCVR from the cell and a second mRNA
encoding a DCVR from the cell; and b) maintaining the isolated cell reaction site, e.g., the cell
isolation micro-chamber, under conditions that allow lysis of the cell and binding of the capture
substrate with the first mRNA and the second mRNA to form the mRNA loaded capture substrate,
wherein the ed cell reaction site, e.g., cell isolation micro-chamber, does not include a nucleic
acid encoding a GCVR or a DCVR from a cell other than the cell (e.g., a different cell).
In an embodiment, the isolated cell reaction site, e.g., cell isolation micro-chamber, comprises
a lysing reagent, e.g., a detergent. In an embodiment, the cell is lysed by heat or an enzyme. In an
embodiment, the capture substrate comprises a moiety (e.g., an oligonucleotide) which binds mRNA,
e. g., an oligo(dT).
In an embodiment, the method further comprises releasing the mRNA loaded capture
substrate from the isolated cell reaction site, e.g., the cell isolation micro-chamber. In an
embodiment, the releasing step is med in the presence of a poly(dA) or poly(dT)
oligonucleotide, e.g., to reduce cross-binding of non-captured mRNA.
In an embodiment, the mRNA loaded capture substrate is transferred from the isolated cell
reaction site, e. g., the cell isolation micro-chamber, to the isolated production reaction site, e.g., the
production micro-chamber.
In an embodiment, the method further ses releasing the nucleic acid sequence from the
isolated production reaction site, e.g., the production micro-chamber. In an embodiment, the method
further comprises amplifying the nucleic acid sequence. In an ment, amplification of the
nucleic acid sequence occurs outside the isolated production reaction site, e.g., the production micro-
chamber, e. g., after the nucleic acid is released from the isolated production reaction site, 6.55., the
production micro-chamber. In an embodiment, ication of the c acid ce occurs at
the isolated production reaction site, e.g., the production micro-chamber.
In an embodiment, the method further comprises sequencing all or a portion of the nucleic
acid sequence.
In an embodiment, the method further comprises inserting all or a portion of nucleic acid
sequence into a vector. In an embodiment, the vector supplies an additional GC element or DC
element not included in the c acid sequence. In an embodiment, the method further ses
expressing the vector.
In an embodiment, the method further comprises expressing the nucleic acid ce to
produce a polypeptide comprising a segment that encodes a GC element of the GCVR, e.g., a
GCVRS, and a segment that encodes a DC element of the DCVR, e. g., a DCVRS. In an embodiment,
the DC element is N-terminal to the GC element in the polypeptide. In an embodiment, the GC
element is C-terminal to the DC element in the ptide.
In an embodiment, the method further comprises contacting the polypeptide with an antigen.
In an embodiment, the method further comprises determining if the ptide binds the antigen, in
vitro, ex vivo, or in viva, e.g., by a method or assay described herein.
In an embodiment, the disclosure features a method of making a nucleic acid sequence
comprising a sequence that encodes a TCR 7 chain element (GC element) of TCR 7 chain variable
region (GCVR) and a TCR 5 chain element (DC element) of a TCR 5 chain variable region (DCVR),
and wherein the GCVR and DCVR are matched, comprising:
a) acquiring an isolated cell reaction site (e.g., an isolated cell reaction site described herein),
e.g., a cell isolation micro-chamber, comprising: i) a cell (e.g., a cell described herein); and ii) a
capture ate (e.g., a capture substrate described herein) capable of binding a first mRNA
encoding a GCVR from the cell and a second mRNA ng a DCVR from the cell;
b) maintaining the isolated cell reaction site, e.g., the cell ion micro-chamber, under
conditions that allow lysis of the cell and binding of the e substrate with the first mRNA and the
second mRNA to form an mRNA loaded capture substrate,
wherein the ed cell reaction site, e.g., cell isolation micro-chamber, does not include a
nucleic acid encoding a GCVR or a DCVR from a cell other than the cell (e.g., a different cell);
c) contacting the mRNA loaded e substrate with a reaction mixture, e.g., a reaction
mixture comprising reverse transcriptase, that uses the loaded mRNA as a template to make cDNA
(this can occur, e.g., in the ed cell reaction site, in an isolated production reaction site, or in
neither, e.g., not in an isolated reaction site);
d) acquiring an isolated production reaction site (e.g., an ed production reaction site
described herein), e.g., a production micro-chamber, comprising: i) a TCR 7 chain (GC) strand,
wherein the GC strand is a strand of a TCR 7 chain double-stranded cDNA (GC ds cDNA) sing
a segment that encodes a GC element of the GCVR from the cell, e.g., a TCR V chain variable region
sequence ); and ii) a TCR 5 chain (DC) strand, n the DC strand is a strand of a TCR 5
chain double-stranded cDNA (DC ds cDNA) comprising a t that encodes a DC element of the
DCVR from the cell, e. g., a TCR 5 chain variable region sequence (DCVRS),
wherein the isolated tion reaction site, e.g., a production micro-chamber, does not
include a nucleic acid encoding a GCVR or a DCVR from a cell other than the cell (e.g., a different
cell); and
e) covalent linking, e.g., ligation, of the GC strand to the DC strand.
In an embodiment, one or more (e.g., two, three, four, or all) of the steps a)-e) are performed
in accordance with a method described herein. In an embodiment, each of the steps a)-e) is performed
in ance with a method described herein.
In an aspect, the sure es a method of making a nucleic acid sequence comprising a
sequence that encodes a TCR V chain element (GC element) of a TCR 7 chain variable region
(GCVR) and a TCR 5 chain element (DC element) of a TCR 5 chain variable region (DCVR), and
wherein the GCVR and DCVR are matched, comprising:
a) acquiring an ed cell reaction site (e.g., an isolated cell reaction site described ),
e.g., a cell isolation micro-chamber, comprising: i) a cell (e.g., a cell described herein); and ii) a
capture substrate (e.g., a capture ate described herein) capable of binding a first mRNA
encoding a GCVR from the cell and a second mRNA encoding a DCVR from the cell;
b) maintaining the isolated cell reaction site, e.g., the cell isolation micro-chamber, under
conditions that allow lysis of the cell and binding of the capture ate with the first mRNA and the
second mRNA to form the mRNA loaded capture substrate,
wherein the isolated cell reaction site, e.g., cell isolation micro-chamber, does not include a
nucleic acid encoding a GCVR or a DCVR from a cell other than the cell (e.g., a different cell);
c) acquiring an isolated production reaction site (e.g., an isolated tion reaction site
described ), e.g., a production micro-chamber, comprises: contacting the mRNA loaded capture
substrate with a on mixture, e.g., a reaction mixture comprising reverse transcriptase, that uses
the loaded mRNA as a template, to produce: a first double-stranded cDNA (ds cDNA) comprising a
strand that is complementary to a first mRNA that encodes a GCVR from a cell; and a second ds
cDNA comprising a strand complementary to a second mRNA encoding a DCVR from the cell (the
cDNA loaded capture substrate);
wherein the isolated production reaction site, e.g., a production micro-chamber, does not
include a nucleic acid encoding a GCVR or a DCVR from a cell other than the cell (e.g., a different
cell).
d) maintaining the isolated production reaction site, e.g., the production micro-chamber,
under conditions that allow amplification of the first and second ds cDNAs, to produce: a plurality of
GC ds cDNAs comprising a segment that encodes a GC element of the GCVR from the cell, e.g., a
GCVRS; and a ity of DC ds cDNAs comprising a segment that encodes a DC element of the
DCVR from the cell, e.g., a DCVRS;
e) acquiring an isolated linkage reaction site (e.g., an isolated linkage on site described
herein), e.g., a e micro-chamber, comprising: covalent linking, e.g., on, of a strand of the
GC ds cDNA (GC strand) to a strand of the DC ds cDNA (DC strand), wherein the GC and DC
strands are both sense s or antisense s; and
f) amplifying the covalently linked, e.g., ligated, GC and DC strands.
In an embodiment, one or more (e.g., two, three, four, five, or all) of the steps a)-f) are
performed in accordance with a method described . In an embodiment, each of the steps a)-f) is
performed in accordance with a method described herein.
WO 19402
In an aspect, the disclosure features a method of making a library comprising a plurality of
unique members, the method comprising:
making the plurality of s, wherein each of the members comprises a sequence that
encodes a TCR V chain element (GC element) of a TCR y chain variable region (GCVR) and a TCR 6
chain element (DC element) of a TCR 5 chain variable region (DCVR), and n the GCVR and
DCVR are matched, made by a method described herein,
wherein each unique c acid sequence of the plurality comprises a GC element and a DC
t from a different unique cell (e.g., a cell described herein),
thereby making a library comprising a ity of unique s.
In an embodiment, the plurality of unique members comprises at least 104, 105, 106, 107, 108,
or 109 unique members. In an embodiment, the plurality of unique members comprises 104 to 109, 104
to 108, 104 to 107, 104 to 106, 104 to 105, 108 to 109, 107 to 109, 106 to 109, 105 to 109, 105 to 108, 106 to
107, 104 to 105, 105 to 106, 106 to 107, 107 to 108, or 108 to 109 unique members. In an embodiment, at
least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%, of the members in the library are unique
members (which encode matched GC element and DC element sequences). In an embodiment, less
than 20%, 15%, 10%, 5%, 4%, 3%, 2%, or 1%, of the members in the library are unique members
(which encode matched GC element and DC element sequences).
In an aspect, the disclosure features a library comprising a plurality of unique members,
wherein,
i) each unique member of the plurality comprises a segment that encodes a GC element, e. g.,
a GCVRS, and a segment that s a DC element, e.g., a DCVRS, n the GC element and
the DC element in each unique member is matched;
ii) each unique member of the plurality comprises a segment that encodes an GC element,
e.g., a GCVRS, and a segment that encodes a DC element, e.g., a DCVRS, from a ent unique
cell; and
iii) the library comprises one or more of the following properties:
a) the library is made by a method described herein;
b) the plurality of unique members comprises at least 104, 105, 106, 107, 108, or 109 unique
nucleic acid sequences;
c) the plurality of unique members comprises 104 to 109, 104 to 108, 104 to 107, 104 to 106, 104
to 105, 108 to 109, 107 to 109, 106 to 109, 105 to 109, 105 to 103, 106 to 107, 104 to 105, 105 to 106, 106 to
107, 107 to 108, or 108 to 109 unique s;
d) at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%, of the members in the
library are unique members (which encode d GC element and DC element sequences); or
e) less than 20%, 15%, 10%, 5%, 4%, 3%, 2%, or 1%, of the members in the library are
unique members (which encode matched GC element and DC element sequences).
In an embodiment, each unique member of the plurality is configured such that, when
expressed, the GC element, e.g., the GCVRS, and the DC element, e.g., the DCVRS, form a
functional antigen binding molecule, e.g., a single chain or a x of a TCR 7 chain and a 5 chain.
In an embodiment, the library is a display library. In an embodiment, each of the s of
the plurality further s a polypeptide that results in display of the member on the e of a
display entity. In an embodiment, the library is a phage y library. In an ment, the
library is a yeast y library. In an embodiment, the library is a mammalian display library.
In an , the disclosure features a method of making a binding polypeptide (e.g., a
ptide sing a GC t and a DC t), the method comprising: a) acquiring a
library described herein, e.g., by a method described herein; and b) expressing a polypeptide encoded
by a unique nucleic acid of the library.
In an embodiment, the method further comprises contacting the polypeptide with an antigen.
In an embodiment, the method further comprises retrieving (e.g., isolating or purifying) the nucleic
acid that encodes a polypeptide that binds the antigen.
In an aspect, the disclosure features an isolated production on site, e.g., a production
micro-chamber, which is an isolated production reaction site described herein (e.g., comprising a
nucleic acid encoding a GCVR and a nucleic acid encoding a DCVR, wherein the GCVR and the
DCVR are matched).
In an embodiment, the isolated production reaction site, e.g., a production micro-chamber,
does not e a nucleic acid encoding a GCVR or a DCVR from a different cell.
In an embodiment, the isolated production reaction site, e.g., a production micro-chamber,
comprises one, two, or all of: (i) one or more primers specific to V gene sequences of the GC and DC;
(ii) one or more primers specific to overhangs introduced onto the GC and DC cDNAs; or (iii) one or
more s comprising a first member, a second member, and a third member comprising a
nucleotide ation (e.g., a spacer) located between the first and second members, wherein the
first member is capable of annealing with the second member of the same primer or a different
primer, e.g., forming a structure comprising a duplex region of 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15,
16, 17, 18, 19, 20, more basepairs.
In an embodiment, the ed production reaction site, e.g., a production micro-chamber,
does not comprise a reagent that can covalently link nucleic acids, e.g., a ligase, e.g., a thermostable
1i gase. In another embodiment, the isolated production reaction site, e. g., a production micro-
chamber, comprises a reagent that can covalently link nucleic acids, e.g., a ligase, e.g., a thermostable
ligase.
In an aspect, the sure features a self-annealing oligonucleotide comprising a first
member, a second , and third member comprising a nucleotide modification (e. g., a spacer)
located between the first and second members, wherein the first member is capable of annealing with
the second member of the same oligonucleotide (e.g., for a method of making a nucleic acid ce
comprising a sequence that encodes a GC element of a GCVR and a DC element of a DCVR, wherein
the GCVR and DCVR are matched).
In an embodiment, the first and second s are capable of forming a hairpin structure
comprising a duplex region of 4, 5,6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, more
irs. In an embodiment, the first member is 5-40 nucleotides, e.g., 5-10, 5-20, 5-30, 30-40, 20-
40, 10-30, 10-30, or 15-25 nucleotides, in length. In an embodiment, the second member is 5-40
nucleotides, e.g., 5-10, 5-20, 5-30, 30-40, 20-40, 10-30, 10-30, or 15-25 nucleotides, in length.
In an ment, the spacer is a spacer described herein, e.g., a flexible spacer or a PEG
spacer.
In an embodiment, the first member comprises a sequence that is complementary to the
sequence of an oligonucleotide attached to a capture substrate.
In an embodiment, the second member comprises (e.g., from 5’ to 3’) one, two, or all of: (i) a
ce that is complementary to at least a portion of the first member; (ii) a sal priming
sequence (e.g., for PCR amplification or next-generation sequencing); and (iii) a sequence
complementary to a target sequence, e.g., a GCVRS and/or a DCVRS. In an embodiment, the
universal priming sequence is identical, or substantially identical, to the sequence that is
complementary to at least a portion of the first member. In another embodiment, the universal
priming sequence is different from the sequence that is complementary to at least a portion of the first
member. In an embodiment, the second member comprises a ce for homologous
recombination (e.g., in a yeast or mammalian cell).
In an aspect, the disclosure features an isolated linkage reaction site, e.g., a linkage micro-
chamber, which is an isolated linkage reaction site described herein (e.g., comprising a nucleic acid
encoding a GCVR and a nucleic acid encoding a DCVR, n the GCVR and the DCVR are
matched).
In an embodiment, the isolated linkage reaction site, e.g., a linkage chamber, does not
include a nucleic acid encoding a GCVR or a DCVR from a different cell.
In an embodiment, the isolated linkage reaction site, 6. g., a linkage micro-chamber, ses
a splint oligonucleotide (e.g., a splint oligonucleotide described herein) that is capable of hybridizing
to a sequence comprising the junction of the GC strand and the DC strand, or a sequence
mentary thereof, to form a duplexed region at the site of ligation.
In an embodiment, the ed linkage reaction site, e.g., a linkage micro-chamber, comprises
a reagent that can covalently link nucleic acids, e.g., a ligase, e.g., a thermostable ligase.
BRIEF DESCRIPTION OF THE GS
s a number of ways of making nucleic acid sequence comprising a ce that
encodes a heavy chain element (HC element) of an antibody heavy chain le region (HCVR) and
a light chain element (LC element) of an antibody light chain variable region (LCVR), and wherein
the HCVR and LCVR are matched. The A1, B1 and C2 boxes indicate steps occurring in an isolated
reaction site, particularly, in an isolated cell reaction site. The C3, D1, D2, D3, D4, D5 and D6 boxes
indicate steps occurring in an isolated reaction site, particularly, in an isolated tion on
site. The E1, E2 and E3 boxes te steps occurring in an isolated reaction site, particularly, in an
isolated linkage reaction site. The C1 box indicates steps that need not occur in an isolated reaction
site. As is discussed in the text, the isolated reaction sites are free of nucleic acid that would result in
a mismatched HC and LC element.
FIGS. 2A-2D are a series of diagrams showing an ary method of making a nucleic acid
comprising a sequence that encodes a heavy chain element (HC element) of an dy heavy chain
variable region (HCVR) and a light chain element (LC element) of an antibody light chain variable
region (LCVR), and wherein the HCVR and LCVR are d. In , a cell (e.g., an immune
cell, such as a B cell) is lysed and mRNAs encoding an HCVR and a matched LCVR are captured on
a bead. In , captured mRNAs are converted to cDNA by reverse transcription followed by
amplification by DNA polymerase (PCR) to create cDNA beads comprising matched pairs of HCVR
and LCVR cDNAs. A self-annealing primer (e.g., a primer comprising a first member and a second
member capable of hybridizing to the first member, with the first and second members separated by a
spacer, e.g., a PEG spacer, and r comprising a ce capable of hybridizing to an HCVR or
LCVR sequence) can be used for the reverse transcription reaction and/or DNA polymerase
amplification. In , matched LCVR and HCVR cDNA products can be fused using a ligase
cycling reaction, in which matched pairs of LCVRs and HCVRs are brought together using a splint
oligo comprising sequences capable of hybridizing to an end of each of the LCVR or HCVR
sequences (e.g., the 3’ terminus of the LCVR and the 5’ terminus of the HCVR). In , the
fused LCVR/HCVR product can be amplified, e.g., by PCR.
is a polyacrylamide gel electrophoresis (PAGE) image showing that Taq ligase and Ampligase
thermostable ligase (Amp; Lucigen) were capable of efficiently linking VH and VL product.
FIGS. 4A-4B are gel electrophoresis images showing that natively paired, linked VH + VL products
for each of antibodies 4G2 and 9E10 were successfully produced by ligase cycling. In ,
denaturing PAGE of ligase cycling products showed that ligase-containing reactions yielded the
linked VH + VL products for each of 4G2 and 9E10, as well as the dual VH and VL
polynucleotides. The linked VH + VL products were not detected in reactions lacking ligase. In 17/068204
4B, agarose gel ophoresis of bulk PCR re-amplification products showed that native pairing was
ed for when VH-VL linked polynucleotides for 4G2 and 9E10 were mixed in the PCR reaction.
FIGS. 5A-SB are a graph and diagram showing efficient and specific PCR product capture using a
nnealing primer. In , a series of forward PCR primer designs were tested for their
capacity to capture PCR t, including (1) a VL primer comprising a spacer and with 5’ sequence
complementary to oligo on bead and 3’ sequence that is complementary to VL template sequence, (2)
a VL primer lacking a spacer and with 5’ sequence complementary to oligo on bead and 3’ sequence
that is complementary to VL template sequence, (3) a VL primer lacking 5’ sequence complementary
to oligo on bead and 3’ sequence that is complementary to VL template ce, and (4) a VH
primer with similar design as in (l) but with 3’-end having sequence complementary to VH template
(for DNA rase extension). In , the VL primer sing a spacer was used for
ent and specific PCR capture of VL oligo, VH oligo, and VH+VL oligo. Of the remaining
primers, only the VH primer was capable of capturing any of the oligos (specifically, the VH oligo
and VH+VL oligo).
is an agarose gel electrophoresis image showing that natively paired VH-VL products could be
produced in drops from nucleic acids obtained from cells expressing the 4G2 antibody. NTC = sample
in which the entire droplet workflow was performed but no cells were included; PCR NTC = no-
template control.
FIGS. 7A-7B are a series of graphs showing that self-annealing primers (in ) can prevent
PCR product e competition at high levels of unused , whereas non-self—annealing primers
(in ) can only do so at low levels of unused primer.
DETAILED DESCRIPTION
Disclosed herein are polypeptides (e.g., antibody molecules or T cell receptor molecules) that
bind to a target molecule or cell, e.g., a human protein or cell, with high affinity and specificity. In an
embodiment, the polypeptide is a binding polypeptide. In an embodiment, the binding polypeptide is
an antibody molecule. In an embodiment, the binding polypeptide is a TCR molecule (e.g., a soluble
TCR molecule). In an embodiment, libraries of the polypeptides, methods for making the
polypeptides or libraries, nucleic acid molecules encoding the polypeptides, expression vectors, host
cells, and compositions (e.g., ceutical compositions), kits, containers, are also provided. The
methods described herein are useful for making or selecting functional polypeptides that contain two
or more chains that are naturally matched or paired. The polypeptides (e.g., antibody les or T
cell or molecules) disclosed herein can be used (alone or in combination with other agents or
therapeutic modalities) to treat, prevent and/or diagnose ers, such as disorders and conditions
disclosed herein.
Without wishing to be bound by theory, it is believed that the methods described herein can
facilitate, e. g., high-throughput phenotypic (e.g., binding) screening of millions of B-cellfplasma cell
antibodies, and antibody discovery from s derived from different species, including, but not
limited to, human, mouse, rat, rabbit, or chicken. For example, the only requirement can be
knowledge of primers to appropriately amplify VH and VL sequences from that species.
Since the workflow described herein is amenable to use in any species, it can significantly
improve ability to er diverse g polypeptides (e. g., antibodies) to target antigens (post
immunization/vaccination), as each species develops different types of binding ptides (e.g.,
antibodies) to an antigen. Immune tolerance issues (e.g., to a target epitope) can be better overcome
by using a species which lacks the target antigen or has significant amino acid differences to the target
antigen, e.g., chicken has reduced tolerance to human antigens/epitopes than human or mouse does to
human antigens/epitopes.
The methods bed herein can facilitate making a ‘phenotypic copy’ of an antibody
repertoire in yeast, which are rugged and can be regrown. This facilitates us and repeated
testing of the dy repertoire, unlike when using primary B-cells, which are sensitive, do no
survive long in vitro, and cannot survive us antibody/BCR binding characterizations.
Other methods to generate natively paired VH-VL ces in droplets can use splicing by
overlap extension with DNA polymerase (PCR) to link the DNA, which may have limitations with
specificity and can result in heterogeneous products of divergent sizes due to imprecise linking. The
ligation methods described here do not suffer from such issues.
Additionally, droplet methods using splicing by overlap extension PCR suffer from an
inherent tion in which any PCR products not fused within drops have the potential to become
fused during non-drop PCR amplification due to the common appended sequence between VH and
VL. Fusion occurring outside of drops leads to non-native pairing, as chains are not
compartmentalized. For the exemplary ligation workflow described herein, there is no need to add
common sequence to VH and VL, and therefore this issue is precluded from occurring.
Such PCR amplification can lead to significantly biased representation of divergent
sequences, as some ces amplify more efficiently than others, which can lead to dramatic
differences after the exponential amplification which occurs in PCR. The workflow described herein
reduces this issue by having PCR products captured onto a bead. For example, if a cell’s VH and VL
sequences are amplified very well or poorly, a similar amount of t will be captured onto the
bead. Thereby, there is a more even representation of dy sequences in the final library, relative
to methods that omit this step and perform linking by splicing by overlap extension PCR.
Definitions
An “HC variable region,” as that term is used , refers to a polypeptide sing
heavy chain CDRs 1, 2 and 3 and heavy chain FW regions 1, 2, 3, and 4.
An “LC variable region,” as that term is used herein, refers to a polypeptide comprising light
chain CDRs 1, 2 and 3 and light chain FW regions 1, 2, 3, and 4.
A “heavy chain variable region sequence,” or “HCVRS,” as that term is used herein, refers to
a ptide comprising ent sequence from heavy chain CDRs and sufficient ce from
heavy chain FW regions, to allow binding of antigen. In embodiments the HCVRS can le with
a light chain variable region, and, e.g., bind antigen. In an embodiment, a HCVRS comprises
sufficient sequence from heavy chain CDRs 1, 2, and 3,and sufficient sequence from heavy chain FW
s, e.g., heavy chain FW regions 1, 2, 3, and 4, to allow binding of antigen. In an embodiment, a
HCVRS comprises heavy chain CDRs 1, 2, and 3, and sufficient sequence from heavy chain FW
regions, e.g., heavy chain FW regions 1, 2, 3, and 4, to complex with a light chain variable region and
to allow binding of antigen.
A “light chain variable region sequence,” or “LCVRS,” as that term is used herein, refers to a
polypeptide comprising ient sequence from light chain CDRs and sufficient sequence from light
chain FW regions, to allow binding of antigen. In embodiments the LCVRS can assemble with a
heavy chain variable region, and, e.g., bind antigen. In an ment, a LCVRS comprises
sufficient ce from light chain CDRs l, 2, and 3,and sufficient sequence from light chain FW
regions, e.g., light chain FW regions 1, 2, 3, and 4, to allow binding of antigen. In an ment, a
LCVRS comprises light chain CDRs 1, 2, and 3, and sufficient sequence from light chain FW regions,
e.g., light chain FW s 1, 2, 3, and 4, to complex with a heavy chain variable region and to allow
binding of antigen.
“Element” of an LC or HC variable region, as that term is used herein, refers to a ce
that encodes at least one amino acid. In an embodiment, an element ses a CDR. In an
embodiment an element comprises a FW region. In an embodiment, and element comprises a CDR
and a FW region. In an embodiment an element comprises a HCVRS or a LCVRS. In an
embodiment, the element comprises at least 10, 20, 30, 40, 50, 60, 70, 80, 90, or 100 amino acid
residues.
A “micro-chamber,” as that term is used herein, refers to a compartment that is dimensioned,
e.g., is sufficiently small, such that upon formation it contains a single cell, or the content from a
single cell. In an ment, the micro-chamber has a volume of that is 10 to 10,000 times greater
of a cell that it contains. In an embodiment, the micro-chamber has a volume of 20 pL. In an
embodiment, the micro-chamber has a maximum dimension of 100 nL. In an embodiment the micro-
chamber comprises a droplet of liquid. In embodiment, the micro-chamber comprises a droplet of a
first liquid disposed in an immiscible media, e.g., a gas or second liquid. In an embodiment, the
micro-chamber comprises a droplet of a first liquid, e. g., a lysis buffer or a PCR reaction buffer,
formed by dispersing the first liquid in an immiscible second liquid, e.g., a fluorinated oil. In an
embodiment, the micro-chamber ses a substrate and a substance other than the substrate, e.g., a
solution. In an embodiment, the droplet comprises a substrate (e.g., a capture substrate, e.g., a bead)
and a substance other than the substrate, e.g., a solution.
“Acquiring,” as that term is used herein, refers to possession of provision of an entity, e.g., a
physical entity or data. Acquiring a physical entity includes making or manufacturing a physical
entity (directly acquiring) as well as ing a physical entity from another party or source
(indirectly acquiring). Acquiring a data or a value includes generating the data or value (directly
acquiring) as well as receiving the data or value from another party or source (indirectly acquiring).
ed,” as that term is used herein in connection with a heavy chain variable region and a
light chain le , means they are from the same cell. With respect to an element of a light
chain variable region and an t of a heavy chain le region it means that the light chain
variable region and the heavy chain variable region from which the elements are derived are from the
same cell.
An “isolated on site,” as that term is used here, refers to a site, e.g., a location on a
substrate, a micro chamber, or a well on a substrate, which allows for sufficient separation between a
first loaded capture substrate and a second loaded capture substrate, or generally, from HC or LC (or
at chain or [3 chain, or 7 chain or 6 chain) encoding nucleic acid of another cell, such that the first
loaded capture substrate is not contaminated with nucleic acid encoding a HC or LC (or 0t chain or [5
chain, or y chain or 5 chain) from another cell. In an embodiment an isolated reaction site provides
sufficient separation between a first mRNA loaded capture substrate and a second mRNA loaded
capture substrate, or generally, from LC or HC encoding c acid of another cell, that the first
loaded mRNA capture substrate is not contaminated with nucleic acid, e. g., mRNA, encoding an HC
or LC (or a chain or [3 chain, or 7 chain or 6 chain) from another cell. In an embodiment an isolated
reaction site provides sufficient separation between a first cDNA loaded capture substrate and a
second cDNA loaded capture substrate, or generally, from HC or LC (or 0. chain or [3 chain, or 7 chain
or 8 chain) encoding nucleic acid of another cell, that the first loaded cDNA e substrate is not
contaminated with nucleic acid, e.g., cDNA, encoding a HC or LC (or 0L chain or [3 chain, or 7 chain or
5 chain) from another cell. Separation can be provided, e.g., by sufficient ce between isolated
reaction sites on a substrate; by configuring the isolated reaction sites such that they are not in fluid
connection, or by formation of an immiscible r between a volume or chamber and the
environment. In an embodiment, the isolated reaction site ses a substrate and a substance other
than the substrate, e.g., a solution.
“Complimentary,” as that term is used herein, refers to sequences which can form Watson-
Crick pairing. When a first sequence is complementary with a second sequence it can be
complementary to the entire second sequence or to less than all of the second sequence.
A “display ,” as that term is used , refers to an entity, e. g., a phage or cell, e. g., a
yeast cell, which includes a gene that encodes a polypeptide.
An “AC variable region,” as that term is used herein, refers to a polypeptide comprising TCR
0t chain CDRs l, 2 and 3 and 0t chain FW regions 1, 2, 3, and 4.
A “BC le region,” as that term is used , refers to a polypeptide comprising [3
chain CDRs 1, 2 and 3 and [3 chain FW regions 1, 2, 3, and 4.
A “GC variable ,” as that term is used herein, refers to a polypeptide comprising TCR V
chain CDRs 1, 2 and 3 and 7 chain FW regions 1, 2, 3, and 4.
A “DC variable region,” as that term is used , refers to a polypeptide comprising 5
chain CDRs l, 2 and 3 and 5 chain FW regions 1, 2, 3, and 4.
An “(1 chain variable region sequence,” or “ACVRS,” as that term is used herein, refers to a
polypeptide sing sufficient sequence from 0t chain CDRs and sufficient sequence from or chain
FW regions, to allow binding of antigen. In embodiments the ACVRS can assemble with a [3 chain
variable region, and, e.g., bind antigen. In an ment, a ACVRS comprises sufficient sequence
from or chain CDRs 1, 2, and 3, and sufficient sequence from or chain FW regions, e.g., or chain FW
regions 1, 2, 3, and 4, to allow g of antigen. In an embodiment, an ACVRS comprises or chain
CDRs 1, 2, and 3, and sufficient sequence from or chain FW regions, e.g., or chain FW regions 1, 2, 3,
and 4, to complex with a [3 chain variable region and to allow binding of antigen.
A “[3 chain variable region sequence,” or ,” as that term is used herein, refers to a
polypeptide comprising sufficient sequence from [3 chain CDRs and sufficient sequence from [3 chain
FW regions, to allow binding of antigen. In embodiments the BCVRS can assemble with an or chain
variable region, and, e.g., bind antigen. In an embodiment, a BCVRS comprises ent sequence
from [3 chain CDRs 1, 2, and 3, and sufficient sequence from [5 chain FW regions, e.g., [3 chain FW
regions 1, 2, 3, and 4, to allow binding of antigen. In an embodiment, a BCVRS comprises [3 chain
CDRs 1, 2, and 3, and sufficient sequence from [3 chain FW s, e.g., [3 chain FW regions 1, 2, 3,
and 4, to complex with an a chain variable region and to allow g of antigen.
A “7 chain variable region sequence,” or “GCVRS,” as that term is used herein, refers to a
polypeptide comprising sufficient sequence from 7 chain CDRs and sufficient sequence from 7 chain
FW regions, to allow binding of antigen. In embodiments the GCVRS can assemble with a 5 chain
variable region, and, e.g., bind antigen. In an embodiment, a GCVRS comprises sufficient sequence
from 7 chain CDRs 1, 2, and 3, and sufficient sequence from y chain FW regions, e.g., y chain FW
regions 1, 2, 3, and 4, to allow binding of antigen. In an embodiment, a GCVRS comprises 7 chain
CDRs l, 2, and 3, and sufficient sequence from y chain FW regions, e.g., 7 chain FW regions 1, 2, 3,
and 4, to complex with a 5 chain variable region and to allow binding of antigen.
A “5 chain variable region sequence,” or “DCVRS,” as that term is used , refers to a
polypeptide comprising sufficient sequence from 5 chain CDRs and ent sequence from 5 chain
FW regions, to allow g of antigen. In embodiments the DCVRS can assemble with a y chain
variable region, and, e.g., bind n. In an embodiment, a DCVRS comprises sufficient sequence
from 5 chain CDRs 1, 2, and 3,and sufficient sequence from 5 chain FW s, e.g., 5 chain FW
regions 1, 2, 3, and 4, to allow binding of antigen. In an embodiment, a DCVRS comprises 5 chain
CDRs 1, 2, and 3, and sufficient sequence from 5 chain FW regions, 6g, 5 chain FW s 1, 2, 3,
and 4, to complex with an or chain variable region and to allow binding of antigen.
“Element” of an 0. chain or [3 chain variable region, as that term is used herein, refers to a
sequence that encodes at least one amino acid. In an embodiment, an element comprises a CDR. In
an embodiment an element comprises a FW region. In an embodiment, and element comprises a
CDR and a FW region. In an embodiment an element comprises an ACVRS or a BCVRS. In an
ment, the element comprises at least 10, 20, 30, 40, 50, 60, 70, 80, 90, or 100 amino acid
residues.
“Element” of a 7 chain or 5 chain variable , as that term is used herein, refers to a
sequence that encodes at least one amino acid. In an embodiment, an t comprises a CDR. In
an embodiment an element comprises a FW region. In an ment, and element comprises a
CDR and a FW region. In an embodiment an element comprises a GCVRS or a DCVRS. In an
embodiment, the t comprises at least 10, 20, 30, 40, 50, 60, 70, 80, 90, or 100 amino acid
residues.
“Matched,” as that term is used herein in tion with an or chain variable region and a [5
chain variable region, means they are from the same cell. With respect to an element of an 0. chain
variable region and an element of a [3 chain variable region it means that the or chain variable region
and the [3 chain variable region from which the elements are derived are from the same cell.
“Matched,” as that term is used herein in tion with a 7 chain variable region and a 5
chain variable region, means they are from the same cell. With t to an element of a y chain
variable region and an element of a 5 chain variable region it means that the 7 chain variable region
and the 7 chain variable region from which the elements are derived are from the same cell.
As used herein, the articles “a” and “an” refer to one or to more than one (e.g., to at least one)
of the grammatical object of the article.
The term “or” is used herein to mean, and is used interchangeably with, the term “and/or”,
unless context clearly indicates otherwise.
“About” and ximately” shall lly mean an acceptable degree of error for the
quantity measured given the nature or ion of the measurements. ary degrees of error are
within 20 percent (%), typically, within 10%, and more typically, within 5% of a given value or range
of values.
The compositions and methods disclosed herein encompass polypeptides and nucleic acids
having the sequences specified, or sequences substantially identical or similar thereto, e.g., sequences
at least 85%, 90%, 95% identical or higher to the sequence specified.
In the context of an amino acid sequence, the term “substantially identical” is used herein to
refer to a first amino acid that contains a sufficient or minimum number of amino acid residues that
are i) identical to, or ii) conservative substitutions of d amino acid residues in a second amino
acid sequence such that the first and second amino acid ces can have a common structural
domain and/or common functional activity. For example, amino acid sequences that contain a
common structural domain having at least about 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%,
98% or 99% identity to a reference sequence, e.g., a sequence provided herein.
In the context of tide sequence, the term antially identical” is used herein to refer
to a first nucleic acid sequence that contains a sufficient or minimum number of nucleotides that are
identical to d nucleotides in a second c acid sequence such that the first and second
nucleotide sequences encode a polypeptide having common functional activity, or encode a common
structural polypeptide domain or a common onal polypeptide activity. For example, nucleotide
sequences having at least about 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99%
ty to a reference sequence, e.g., a ce provided herein.
The term “functional variant” refers polypeptides that have a substantially identical amino
acid sequence to the lly-occurring sequence, or are encoded by a substantially identical
nucleotide sequence, and are capable of having one or more activities of the naturally-occurring
sequence.
ations of homology or sequence identity between sequences (the terms are used
interchangeably herein) are performed as s.
To determine the percent identity of two amino acid sequences, or of two nucleic acid
sequences, the ces are aligned for optimal comparison purposes (e.g., gaps can be introduced in
one or both of a first and a second amino acid or nucleic acid sequence for optimal alignment and
non-homologous sequences can be disregarded for comparison purposes). In a typical embodiment,
the length of a reference sequence aligned for comparison purposes is at least 30%, e.g., at least 40%,
50%, 60%, e.g., at least 70%, 80%, 90%, 100% of the length of the reference sequence. The amino
acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then
compared. When a position in the first sequence is occupied by the same amino acid residue or
nucleotide as the corresponding position in the second ce, then the molecules are identical at
that position.
The percent ty between the two sequences is a function of the number of identical
positions shared by the sequences, taking into account the number of gaps, and the length of each gap,
which need to be introduced for optimal alignment of the two sequences.
The comparison of sequences and ination of percent identity between two sequences
can be accomplished using a mathematical algorithm. In some embodiments, the percent identity
n two amino acid sequences is determined using the Needleman and Wunsch ((1970) J. M01.
Biol. 48:444-453) algorithm which has been incorporated into the GAP program in the GCG software
package (available at www.gcg.com), using either a Blossum 62 matrix or a PAM250 matrix, and a
gap weight of 16, 14, 12, 10, 8, 6, or 4 and a length weight of l, 2, 3, 4, 5, or 6. In certain
embodiments, the percent identity between two nucleotide sequences is determined using the GAP
program in the GCG software e (available at www.gcg.com), using a NWSgapdna.CMP matrix
and a gap weight of 40, 50, 60, 70, or 80 and a length weight of l, 2, 3, 4, 5, or 6. One suitable set of
parameters (and the one that should be used unless otherwise ied) are a Blossum 62 scoring
matrix with a gap penalty of 12, a gap extend penalty of 4, and a frameshift gap penalty of 5.
The percent ty between two amino acid or tide sequences can be determined
using the thm of E. Meyers and W. Miller ((1989) , 4: 11-17) which has been
incorporated into the ALIGN program (version 2.0), using a PAM120 weight residue table, a gap
length penalty of 12 and a gap penalty of 4.
The nucleic acid and protein sequences described herein can be used as a “query sequence” to
perform a search against public databases to, for example, identify other family members or related
sequences. Such searches can be performed using the NBLAST and XBLAST programs (version 2.0)
of Altschul, et al. (1990) J. Mol. Biol. 215:403-10. BLAST nucleotide searches can be performed
with the NBLAST program, score = 100, wordlength = 12 to obtain nucleotide sequences homologous
to a nucleic acid as described . BLAST protein searches can be med with the XBLAST
m, score = 50, ngth = 3 to obtain amino acid sequences homologous to protein
molecules described herein. To obtain gapped alignments for comparison purposes, Gapped BLAST
can be utilized as described in Altschul et al., (1997) Nucleic Acids Res. 25 :3389-3402. When
utilizing BLAST and gapped BLAST programs, the default parameters of the respective programs
(e.g., XBLAST and NBLAST) can be used. See www.ncbi.nlm.nih.gov.
As used herein, the term “hybridizes under low stringency, medium stringency, high
stringency, or very high stringency conditions” bes conditions for ization and washing.
Guidance for performing hybridization reactions can be found in Current Protocols in Molecular
Biology, John Wiley & Sons, NY. (1989), 63.1-63.6, which is incorporated by reference. Aqueous
and eous methods are described in that nce and either can be used. Specific
hybridization conditions referred to herein are as follows: 1) low stringency hybridization conditions
in 6X sodium chloride/sodium citrate (SSC) at about 45°C, followed by two washes in 0.2X SSC,
0.1% SDS at least at 50°C (the temperature of the washes can be increased to 55°C for low stringency
conditions); 2) medium stringency hybridization conditions in 6X SSC at about 45°C, followed by
one or more washes in 0.2X SSC, 0.1% SDS at 60°C; 3) high stringency hybridization conditions in
6X SSC at about 45°C, followed by one or more washes in 0.2X SSC, 0.1% SDS at 65°C; and
preferably 4) very high stringency ization conditions are 0.5M sodium ate, 7% SDS at
65°C, followed by one or more washes at 0.2X SSC, 1% SDS at 65°C. Very high stringency
conditions 4) are suitable conditions and the ones that should be used unless otherwise specified.
It is understood that the molecules described herein may have additional conservative or non-
essential amino acid substitutions, which do not have a substantial effect on their functions.
2017/068204
The term “amino acid” is intended to embrace all molecules, whether natural or synthetic,
which include both an amino functionality and an acid functionality and capable of being included in
a polymer of naturally-occurring amino acids. Exemplary amino acids include lly-occurring
amino acids; analogs, derivatives and congeners thereof; amino acid analogs having t side
chains; and all stereoisomers of any of any of the foregoing. As used herein the term “amino acid”
includes both the D- or L- optical isomers and peptidomimetics.
A “conservative amino acid substitution” is one in which the amino acid residue is replaced
with an amino acid residue having a similar side chain. Families of amino acid residues having
similar side chains have been defined in the art. These families include amino acids with basic side
chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid),
uncharged polar side chains (6.3., glycine, asparagine, glutamine, serine, threonine, tyrosine,
cysteine), nonpolar side chains (e. g., e, valine, e, isoleucine, proline, alanine,
methionine, tryptophan), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic
side chains (e.g., tyrosine, phenylalanine, tryptophan, ine).
The terms eptide,93 upeptide” and “protein” (if single chain) are used interchangeably
herein to refer to polymers of amino acids of any length. The polymer may be linear or branched, it
may comprise modified amino acids, and it may be interrupted by non-amino acids. The terms also
encompass an amino acid polymer that has been modified; for example, de bond formation,
glycosylation, lipidation, ation, phosphorylation, or any other manipulation, such as conjugation
with a labeling component. The ptide can be isolated from natural sources, can be a produced
by recombinant techniques from a eukaryotic or prokaryotic host, or can be a product of synthetic
procedures. In an embodiment, the polypeptide is an antibody molecule. In another embodiment, the
polypeptide is a TCR molecule, e.g., soluble TCR molecule.
The terms “nucleic acid,” “nucleic acid sequence,a, unucleotide sequence,” or “polynucleotide
sequence,” and “polynucleotide” are used interchangeably. They refer to a ric form of
nucleotides of any length, either deoxyribonucleotides or ribonucleotides, or s thereof. The
polynucleotide may be either single-stranded or double-stranded, and if single-stranded may be the
coding strand or non-coding (antisense) strand. A polynucleotide may comprise modified
nucleotides, such as methylated nucleotides and nucleotide analogs. The sequence of nucleotides may
be interrupted by cleotide components. A polynucleotide may be further modified after
polymerization, such as by conjugation with a labeling component. The nucleic acid may be a
recombinant polynucleotide, or a polynucleotide of genomic, cDNA, semisynthetic, or synthetic
origin which either does not occur in nature or is linked to another polynucleotide in a non-natural
arrangement.
The term “isolated,” as used herein, refers to material that is removed from its original or
native environment (e.g., the natural environment if it is naturally occurring). For example, a
lly-occurring cleotide or polypeptide present in a living animal is not isolated, but the
same polynucleotide or polypeptide, separated by human intervention from some or all of the co-
ng materials in the natural , is isolated. Such polynucleotides could be part of a vector
andior such polynucleotides or polypeptides could be part of a composition, and still be isolated in
that such vector or composition is not part of the environment in which it is found in nature.
As used herein, the term “treat,” e.g., a disorder described herein, means that a subject (e.g., a
human) who has a disorder, e.g., a disorder described herein, and/or experiences a symptom of a
disorder, e.g., a disorder described herein, will, in an embodiment, suffer less a severe m
and/or recover faster when an antibody molecule is stered than if the antibody molecule were
never administered. Treatment can, e.g., partially or tely, ate, ameliorate, relieve, inhibit,
or reduce the severity of, and/or reduce incidence, and optionally, delay onset of, one or more
manifestations of the effects or symptoms, features, and/or causes of the disorder. In an ment,
treatment is of a subject who does not exhibit certain signs of the disorder, and/or of a subject who
exhibits only early signs of the disorder. In an embodiment, treatment is of a subject who exhibits one
or more established signs of a disorder. In an embodiment, treatment is of a subject diagnosed as
suffering from a disorder.
As used , the term “prevent,” a disorder, means that a subject (e.g., a human) is less
likely to have the er, if the subject receives a polypeptide (e.g., antibody le).
Various aspects of the itions and methods herein are described in further detail below.
Additional definitions are set out hout the specification.
Libraries of Binding Polypeptides
Disclosed herein are libraries (e.g., display libraries) of binding polypeptides, e. g., antibody
molecules or T cell receptor molecules, and methods of making libraries of binding polypeptides, e.g.,
antibody molecules or T cell receptor molecules.
In an embodiment, a method described herein links two DNA fragments, such as sequences
ng an antibody heavy chain variable region (or a portion thereof) and an antibody light chain
variable region (or a portion thereof), a TCR or chain (or a portion thereof) and a TCR [3 chain (or a
portion thereof), or a TCR 7 chain (or a portion f) and a TCR 5 chain (or a portion thereof),
using a ligase-mediated approach.
For example, antibodies are ed of two types of polypeptide chains, light chain and
heavy chain, each of which are translated from separate mRNA molecules. In order to copy a
functional unit of a particular antibody (or B cell receptor) from a B cell, knowledge of the particular
heavy chain and its cognate light chain must be maintained. This is typically performed using
methods in which dual clones are in wells of microwell plates, which keeps clones segregated
and the result heavy and light chain sequences thus are known to be paired. Such cloning processes
scale well to B cell numbers compatible with 96- or ll plates. However, B cell repertoires in
humans and animals can range from about 106-1011 B cells, many of which are different clones (La,
different BCRs or antibodies). Thus, there is a need to be able to, in an efficient manner, make copies
of millions to billions of B cells which (1) retains native pairing of chains and (2) allows for
functional interrogation of such a large number of unique clones. Such a method can tate
making a renewable copy of an dy repertoire which can be functionally interrogated by a
variety of methods.
In an embodiment, a method described herein uses one or more (e.g., two, three, or all) of the
following: (1) miniaturized compartmentalization of individual cells (e.g., B cells or T cells) in
droplets (pL to nL volume drops), (2) lysing and PCR amplifying two chains (e.g., antibody VH and
VL, TCR 0t and [3 chains, or TCR V and 5 chains), (3) ically linking the two , such that
native chain pairing is retained and that a thermostable ligase catalyzes the linking, and (4) amplifying
the linked DNA in a manner that allows for high throughput phenotypic interrogation of clones by a
surface y technology, such as yeast or phage display.
The methods bed herein can result in a nucleic acid sequence, when expressed, encodes
a onal polypeptide, e.g., a functional n binding polypeptide. For example, the HC element
and the LC element (or the AC element and the BC element, or the GC element and the DC element)
are not ured in a head-to-head or tail-to-tail orientation. In an embodiment, the HC element and
the LC t (or the AC element and the BC element, or the GC t and the DC element) are
configured in a head-to-tail orientation. For example, the C-terminus of the LC element (or LCVRS)
is linked, directly or indirectly, with the N-terminus of the HC element (or HCVRS), or the C-
terminus of the HC element (or HCVRS) is linked, directly or indirectly, with the inus of the
LC element (or LCVRS).
Exemplary Workflow
Cells (e.g., immune cells, e.g., B cells or T cells) are encapsulated individually into drops. In
the drops, the cells are lysed and mRNA is captured onto beads, which contain oligonucleotides to
hybridize to mRNA. The beads facilitate maintaining native pairing information (e.g., the native
pairing between two chains, e.g., a heavy chain and a light chain in a single B cell; an 0t chain and a [3
chain in a single T cell; or a 7 chain and a 6 chain in a single T cell). Next, the mRNA is reversed
transcribed to cDNA by a reverse riptase (RT). The reverse transcription can be performed
within the lysis drops, outside of drops, or in the subsequent drop (‘PCR’ drop). Beads having
captured mRNA or cDNA are recovered from the initial drops. The beads are then encapsulated into
new drops, wherein the nucleic acids are amplified, either by RT-PCR (when mRNA is template) or
PCR (when cDNA is template). The cDNAs encoding the two chains are amplified in drops. The
amplified products are captured back onto beads by ic complementary nucleic acid
hybridization. The beads having captured products are recovered from drops and subsequently
encapsulated into new drops. The amplified product encoding one chain (e.g., VH) is linked with the
amplification t encoding the other chain (e. g., VL) in drops using a thermostable ligase. In an
approach (“linking cohesive products”), cohesive (or “sticky-end”) PCR products are generated, and
covalent ligation of hybridized cohesive PCR products are performed by a thermostable ligase. In
another approach se cycling reaction”), no cohesive PCR products are produced. , in
drops, DNA is linked together h use of a thermostable ligase and a splint (or bridging)
ucleotide. While not wishing to be bound by theory, it is believed that in an embodiment, the
methods described herein reduce or preclude the possibility of nded fusing caused by overlap
extension PCR s (Turchaninova et al. Eur J l. 2013; 43(9): 2507-2515). The ligated
products, representing natively paired chains, are further amplified to generate sufficient material to
create a display library, such as in yeast or phage. The amplified product, encoding ly paired
chains (e.g., antibody heavy chain and light chain, TCR 0L chain and [3 chain, or TCR 7 chain and 5
chain) in a format such as an scFv, scFab, Fab, or full-length IgG, are uced to an appropriate
expression or display vehicle, such as yeast or phage display. The constructed library, e. g., having
>104 and up to 109 or larger members, can be rapidly interrogated for desired g and/or other
phenotypic properties, using established methods.
Generation of Cohesive PCR Products That Are Suitable Substratesfor Ligase
In an embodiment, amplification (e.g., PCR) products with cohesive ends that are suitable
substrates for ligase are generated. Without wishing to be bound by theory, it is believed that in an
embodiment, DNA polymerase extension can be prematurely terminated at a defined on, e.g.,
through use of a ally modified (e.g., lesioned) nucleotide or base, or other alterations to the
primer used for amplification. These chemically modified nucleotides or bases (or other primer
alterations) are subsequently incorporated into one strand of the ication product. As the DNA
polymerase reads through the template strand which contains the modified nucleotide, it prematurely
stops extension at (or near) the modified nucleotide, as it is not able to read through. This early
polymerase termination due to the modification can lead to production of an amplification product
with a cohesive end. The amplification product can hybridize (or anneal) efficiently with another
amplification product having a complementary cohesive end, which can be produced in an analogous
matter. For example, a PCR product encoding one chain (6.3., VH) and a PCR t encoding
another chain (e.g., VL), each having a complementary cohesive end, can hybridize (or anneal) to
each other with high ncy. Next, a thermostable ligase, present in the droplet with the DNA
polymerase (e. g., hout thermocycling), catalyzes ligation (covalent linkage) of the hybridized
(or annealed) DNA les.
In an embodiment, the native paring information is maintained during amplification and
ligation. In an embodiment, both amplification and ligation occur in the same drop, e.g., without
breaking the drop. In an embodiment, the ligase retains at least 50%, e.g., at least 60%, 70%, 80%,
85%, 90%, 95%, or 99% ty, at 95°C or more (e.g., 96°C or more, 97°C or more, or 98°C or
more), during one or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, or more) thermocycles. In an
embodiment, the ligase retains at least 50%, e.g., at least 60%, 70%, 80%, 85%, 90%, 95%, or 99%
ty, at 95°C or more (e.g., 96°C or more, 97°C or more, or 98°C or more), for at least 5 minutes
(e. g., at least 10 minutes, 15 minutes, 20 minutes, 25 minutes, 30 minutes, 45 minutes, or 60 minutes).
In an ment, the ligase retains at least 50%, e.g., at least 60%, 70%, 80%, 85%, 90%, 95%, or
99% activity, at 95°C or more (e.g., 96°C or more, 97°C or more, or 98°C or more), in a buffer
ion that allows for DNA polymerase activity.
In an embodiment, the modification inhibits or blocks DNA polymerase activity and s
a substrate for ligation. In an embodiment, the modification does not inhibit or prevent binding of the
amplification product to a ligase. In an embodiment, the modification does not inhibit or prevent
formation of a phosphodiester bond. In an embodiment, the cation does not se a large
bulky chemical group. Exemplary modifications include, but are not limited to, a ribose 2’-C (2nd
carbon) modification (e.g., OH (516., a ribonucleotide, not a deoxyribonucleotide), O-methyl (O-CH3),
or amine (NH2)); a ribose 4’-C (4th carbon) ation; a base modification (e.g., a non-native base,
a uracil, or others); an abasic site (e.g., an AP site or apyrimidine/apurine); or a staggered primer (e.g.,
different length ng). In an embodiment, the modification comprises a uracil and a DNA
polymerase that is inhibited by uracil (e.g., an archaeal DNA polymerase) is used for amplification.
Exemplary steps for performing a cohesive-end PCR-ligation experiment in drops are
illustrated below.
Cell Encapsulation
Cells can be encapsulated individually into droplets. In an embodiment, the cell is an immune
cell. In an embodiment, the cell is a B cell. In an ment, the cell is a T cell. In an embodiment,
the cell is an antibody-producing cell. In an embodiment, the cell is an isolated cell or purified cell.
In an embodiment, the cell is obtained from a subject, e.g., a human, mouse, rabbit, rat, goat, sheep, or
chicken.
In an embodiment, the volume of the droplet is from 10 pL to 100 nL, e.g., from 10 pL to 100
pL, from 10 pL to 1000 pL, from 10 pL to 10 nL, from 10 nL to 100 nL, from 1000 pL to 100 nL,
from 100 pL to 100 nL, from 100 pL to 10 nL, from 100 pL to 1000 pL, from 1000 pL to 10 nL, or
from 100 pL to 1000 pL. In an embodiment, the volume of the droplet is from 100 pL to 1000 pL.
In an embodiment, the droplet is a in-oil droplet. In an embodiment, the droplet is
present in a carrier (e.g., oil) phase, e.g., a carrier phase comprising 3MTM HFE-7500 with about 1%
fiuorosurfactant (RAN Biotechnologies).
The droplets can be formed, e. g., using a microfluidic chip (e.g., 2R 100 pm from Dolomite)
with the flow of fluid phase controlled by a syringe or pressure pump. In an embodiment, the aqueous
phase of the droplet comprises a , a reagent that aids cell lysis, and a bead. In an embodiment,
the buffer comprises Tris at pH 7.5. In an ment, the reagent that aids cell lysis comprises a
detergent. Exemplary detergents that can be used to aid cell lysis include, but are not limited to,
Tween-20, Triton X, IGEPAL, or sodium lauroyl sarcosinate (Sarkosyl). In an embodiment, the bead
is a magnetic bead. In an ment, the bead comprises, is coupled to, an oligonucleotide (e. g., a
primer), e. g., to anneal to an mRNA (e. g., an mRNA encoding a heavy chain or a light chain).
In an embodiment, the droplet contains no more than one cell after encapsulation. In an
embodiment, the droplet contains a plurality of beads. In an embodiment, a plurality of beads are
obtained, and at least 80%, e. g., at least 85%, 90%, 95 %, 98%, 99%, or 100%, of the plurality
contains no more than one cell per droplet. In an embodiment, a plurality of beads are obtained, and
at least 80%, e. g., at least 85%, 90%, 95%, 98%, 99%, or 100%, of the plurality ns at least one
bead per droplet. Typically, the occupancy of drops is no more than one cell per droplet, and at least
one bead per droplet.
Cell Lysis
After encapsulation, the droplets can be ted to tate cell lysis. In an embodiment,
an emulsion (e. g., ning coalesce different solution phases) is heated, e. g., to reduce mRNA
secondary structures so that it can be more efficiently ed onto the bead and/or to improved lysis
efficiency in the presence of a detergent (e. g., Tween20). In an embodiment, the emulsion is
ted at a temperature between 40°C and 80°C, e. g., between 40°C and 60°C, 50°C and 70°C, or
60°C and 80°C, e. g., at 40°C, 50°C, 60°C, 70°C, or 80°C. In an embodiment, the emulsion is
incubated for 5 to 60 minutes, e.g., 10 to 45 minutes, 15 to 30 minutes, 5 to 30 minutes, or 30 to 50
minutes. In an embodiment, the cell is lysed by heat. In an embodiment, the cell is lysed by an
enzyme. Typically, after the cell is lysed, mRNA is released and is captured on a bead by annealing
to the oligonucleotides on the bead.
Bead Recovery
Emulsions (e.g., containing coalesce different solution phases) can be broken using a drop
destabilizing reagent, e.g., perfluorooctanol (PFO). In an embodiment, the bead-containing aqueous
phase is recovered. In an embodiment, the bead is a magnetic bead, and is isolated using magnet. In
an embodiment, the bead is washed and resuspended in a buffer (e. g., Tris, pH 7.5).
Reverse Transcription
Reverse transcription can be performed using standard methods. In an embodiment, the
reverse transcription is performed in a non-emulsion reaction. In an embodiment, the reverse
transcription is performed in an emulsion reaction. In an embodiment, the bead with ed mRNA
is resuspended in a buffer-enzyme mix (e.g., Superscript II RT) and incubated at 35°C to 45°C (e. g.,
at 40°C) for 10 to 60 minutes (e.g., 15 minutes) to facilitate reverse transcription. In an ment,
the oligonucleotide coupled to the bead is used as a primer for the synthesis of the first strand cDNA.
In an embodiment, the bead is washed with a buffer (e.g., Tris, pH 7.5) after e transcription.
Bead Encapsulation
Beads can be encapsulated individually into droplets. In an ment, the volume of the
droplet is from 5 pL to 500 pL, e. g., from 5 pL to 400 pL, from 5 pL to 300 pL, from 5 pL to 200 pL,
from 5 pL to 100 pL, from 5 pL to 50 pL, from 5 pL to 25 pL, from 400 pL to 500 pL, from 300 pL to
500 pL, from 200 pL to 500 pL, from 100 pL to 500 pL, from 50 pL to 500 pL, from 25 pL to 500 pL,
from 10 pL to 500 pL, from 10 pL to 400 pL, from 25 pL to 300 pL, from 50 pL to 200 pL, or from
pL to 50 pL. In an embodiment, the volume of the droplet is from 10 pL to 50 pL.
In an embodiment, the droplet is a water-in-oil droplet. In an embodiment, the droplet is
present in a carrier (e.g., oil) phase, e.g., a carrier phase comprising 3MTM HFE-7500 with about 1%
fluorosurfactant (RAN Biotechnologies).
In an embodiment, the droplet contains one bead after encapsulation. In an embodiment, a
plurality of beads are obtained, and at least 80%, e.g., at least 85%, 90%, 95%, 98%, 99%, or 100%,
of the plurality contains no more than one bead per droplet.
PCR-Ligarion Reaction
In an embodiment, a PCR-ligation reaction is performed. In an embodiment, the PCR-
ligation reaction generates a ligated product (e.g., a double-stranded DNA) that comprises a
nucleotide sequence that encodes an antibody heavy chain variable region (or a portion thereof) and
an antibody light chain variable region (or a portion thereof). In an embodiment, the PCR-ligation
on generates a ligated product (e.g., a -stranded DNA) that ses a tide
sequence that encodes a TCR (1 Chain (or a portion thereof) and a TCR [3 chain (or a portion thereof).
In an embodiment, the PCR-ligation reaction generates a ligated product (e.g., a double-stranded
DNA) that comprises a nucleotide sequence that encodes a TCR 7 chain (or a portion thereof) and a
TCR 5 chain (or a portion thereof). In an ment, the PCR-ligation reaction is performed in a
droplet comprising a bead that is coupled with cDNA, a DNA polymerase, oligonucleotides (e.g., for
amplification of the cDNA), a ligase (e.g., a thermostable ligase), and a buffer.
Exemplary DNA polymerases that can be used in the reaction include, but are not d to,
Phusion® High-Fidelity DNA Polymerase (NEB), Q5® High-Fidelity DNA rase (NEB), Pfu
DNA polymerase, KAPA DNA polymerase, Vent® DNA polymerase, or Taq DNA polymerase.
In an embodiment, the ligated product contains an scFv, a Fab, or scFab cassette. In an
ment, the cassette (e.g., scFv te) is constructed as VL-Linker-VH. Without g to
be bound by theory, it is believed that in an embodiment, the order can be switched to VH-Linker-VL,
with no significant impact on expression or function. In an embodiment, the cassette (e.g., scFv)
cassette is constructed as VH-Linker-VL. In an ment, the cassette comprises a constant region
sequence (e.g., a CH1 domain and/or a CL ), e.g., VH-CHl coupled with VL-CL.
WO 19402
Similarly, the ligated product can contain a cassette constructed as at Linker-B chain or
[3 chain-Linker-a chain, or y chain-Linker-5 chain or 5 chain-Linker-y chain.
In an embodiment, the reverse primer for the VL sequence contains one, two, or all of the
following: (a) an overhang sequence encoding a linker sequence (b) at least one modified tide
(e.g., 3 consecutive nucleotides with ethy1 modification), e.g., in the overhang; or (c) a 5’-
phosphate. In an embodiment, the forward primer for the VH sequence contains one, two, or all of
the following: (a) an overhang sequence encoding a linker sequence (b) at least one modified
nucleotide (e.g., 3 consecutive nucleotides with 2’-O-methyl modification), e.g., in the overhang; or
(c) a 5’-phosphate.
In an embodiment, the reverse primer for the VH sequence contains one, two, or all of the
following: (a) an overhang sequence encoding a linker sequence (b) at least one modified nucleotide
(e. g., 3 utive nucleotides with 2’-O-methyl modification), e.g., in the overhang; or (c) a 5’-
phosphate. In an embodiment, the forward primer for the VL sequence contains one, two, or all of the
following: (a) an overhang ce encoding a linker sequence (b) at least one modified nucleotide
(6.3., 3 consecutive nucleotides with 2’-O-methyl modification), e.g., in the ng; or (c) a 5’-
phosphate.
rly, in an embodiment, the reverse primer for the OL chain (or 7 chain) sequence contains
one, two, or all of the following: (a) an overhang sequence encoding a linker sequence (b) at least one
modified nucleotide (e.g., 3 consecutive nucleotides with 2’-O-methyl modification), e.g., in the
overhang; or (c) a 5’-phosphate. In an embodiment, the forward primer for the B chain (or 6 chain)
sequence contains one, two, or all of the following: (a) an overhang sequence encoding a linker
sequence (b) at least one modified nucleotide (e. g., 3 consecutive nucleotides with 2’-O-methyl
modification), e.g., in the overhang; or (c) a 5’-phosphate.
Similarly, in an ment, the reverse primer for the [3 chain (or 5 chain) sequence contains
one, two, or all of the following: (a) an overhang sequence encoding a linker sequence (b) at least one
modified tide (e.g., 3 consecutive nucleotides with 2’-O-methyl modification), e.g., in the
overhang; or (c) a 5’-phosphate. In an embodiment, the forward primer for the 0: chain (or 7 chain)
sequence contains one, two, or all of the following: (a) an overhang ce encoding a linker
ce (b) at least one modified nucleotide (e.g., 3 consecutive nucleotides with 2’-O-methyl
modification), e.g., in the overhang; or (c) a 5’-phosphate.
ary ligases (e.g., thermostable ligases) that can be used in the on include, but are
not limited to, Taq DNA ligase, Pfu DNA ligase, Ampligase® thermostable DNA ligase, Tsc DNA
ligase, Rma DNA ligase, Tfi DNA ligase, or Tth DNA .
In an embodiment, the buffer supports both DNA polymerase and ligase enzymatic ties.
In an embodiment, the cycling is performed with emulsion (e.g., in a PCR tube). In an
embodiment, the thermocycling is performed using the following conditions: initial denaturation at
95-98°C for 30 seconds to 2 minutes; 10-30 cycles of: denaturation at 95-98°C for 10-30 seconds,
primer annealing at 50-60°C for 10-30 seconds, polymerase extension at 72°C for 30 seconds, and
cohesive product annealing and ligation at C for 3 minutes. The reaction can be hold at 4°C.
Recovery ofAqueous n
Emulsions (e.g., containing coalesce different solution phases) can be broken using a drop
destabilizing reagent, e. g., perfluorooctanol (PFO). In an embodiment, the aqueous portion (e. g.,
containing linked product, and optionally, non-linked product) is recovered. In an embodiment, the
bead is discarded.
Purification ofLinked Product
Linked product (e.g., enting natively linked VL—linker-VH) can be purified from non-
linked products (e. g., non-linked VH and VL). The ligated products are separated from non-ligated
products by size tion. For example, denaturing PAGE (polyacrylamide gel electrophoresis) or
denaturing HPLC-SEC can be used. The linked product (~800-900 bp) is isolated from non-linked
product (~350-500 bp). For denaturing PAGE purification, the d band is cut out from the gel
and an elecro-elution is performed to extract DNA from gel slice (Bio-Rad Electro-Elutor).
Amplification ofPurified Linked Product
The purified linked product can be amplified, e. g., by PCR. For e, the purified linked
product is amplified by PCR using a DNA polymerase (e.g., Taq polymerase) under conditions that
can tely read through DNA containing modified nucleotides.
The final PCR product can be introduced to yeast using rd methods (e.g.,
electroporation with expression vector) to create a natively paired library derived from biological
Ligase Cycling
In an embodiment, the different chains (e. g., VH and VL) are not amplified in a manner
which incorporates DNA sequence common to both chains, which would facilitate ing of
-end ts directly to each other. In an embodiment, a bridging (or splint) oligonucleotide is
ed after the amplification of cDNA but in the presence of a thermostable ligase. The bridging
oligonucleotide can facilitate bringing the two chains immediately adjacent to each other such that
they become a substrate of ligase. Ligase, in turn, catalyzes a covalent bond formation between
chains of DNA. Since this mechanism does not lead to incorporation of sequence common to both
chains in each chain (e.g., overhang DNA with common sequence in both VH and VL), there is no
opportunity for splicing by overlap extension PCR.
The steps of this approach are generally the same as above except beginning at the emulsion
PCR step. PCR amplification of cDNA can be performed in drops. Primers can add overhang
sequences, but there is generally no common sequence to both chains (e.g., VL and VH) that is added
(unlike the above strategy). The bead in the drop, through its conjugated oligonucleotides, becomes
filled or saturated with dsDNA ts of the two chains (e.g., VH and VL), each with specific
overhang ce. Drops are broken, and any PCR product not linked or annealed to beads is
washed away. Beads ning dsDNA of two chains (e.g., VH and VL) are encapsulated into new
drops in the presence of a thermostable ligase and a splint oligonucleotide. In this emulsion format,
thermocycling is performed, which facilitates formation of the 3-DNA piece complex. This complex
is a substrate for ligase, which zes covalent bond formation, linking the two chains. In an
embodiment, thermocycling aids sion of all ‘top ’ DNA to linked product, until one
substrate becomes limiting. In another ment, both strands become ligated. For example, once
the ‘top strand’ is ligated, it can serve as the ‘splint’ for the opposing strand, which ligase will
recognize as a substrate. Without wishing to be bound by theory, it is believed that, this facet
specifically makes the reaction nt, that is, initial ligated product can serve as more templates
(splints) to generate even more additional ligated product. Drops are broken, and the ligated products
are amplified by rd PCR means.
Exemplary steps for performing a ligase cycling experiment are illustrated below.
Cell Encapsulation
Cells can be encapsulated individually into droplets. In an embodiment, the cell is an immune
cell. In an embodiment, the cell is a B cell. In an embodiment, the cell is a T cell. In an embodiment,
the cell is an antibody-producing cell. In an ment, the cell is an isolated cell or purified cell.
In an embodiment, the cell is obtained from a subject, e.g., a human, mouse, rabbit, rat, goat, sheep, or
chicken.
In an embodiment, the volume of the droplet is from 10 pL to 100 nL, e.g., from 10 pL to 100
pL, from 10 pL to 1000 pL, from 10 pL to 10 nL, from 10 nL to 100 nL, from 1000 pL to 100 nL,
from 100 pL to 100 nL, from 100 pL to 10 nL, from 100 pL to 1000 pL, from 1000 pL to 10 nL, or
from 100 pL to 1000 pL. In an embodiment, the volume of the droplet is from 100 pL to 1000 pL.
In an embodiment, the droplet is a water-in-oil droplet. In an embodiment, the droplet is
present in a carrier (e.g., oil) phase, e.g., a carrier phase comprising 3MTM HFE-7500 with about 1%
fluorosurfactant (RAN Biotechnologies).
The droplets can be formed, e.g., using a luidic chip (e.g., 2R 100 pm from Dolomite)
with the flow of fluid phase controlled by a syringe or pressure pump. In an embodiment, the aqueous
phase of the droplet comprises a buffer, a reagent that aids cell lysis, and a bead. In an ment,
the buffer comprises Tris at pH 7.5. In an embodiment, the reagent that aids cell lysis comprises a
detergent. Exemplary detergents that can be used to aid cell lysis include, but are not limited to,
Tween-20, Triton X, , or sodium lauroyl sarcosinate (Sarkosyl). In an embodiment, the bead
WO 19402
is a magnetic bead. In an embodiment, the bead comprises, is coupled to, an oligonucleotide (e. g., a
primer), e. g., to anneal to an mRNA (e. g., an mRNA encoding a heavy chain or a light chain).
In an ment, the droplet contains no more than one cell after encapsulation. In an
embodiment, the droplet contains a plurality of beads. In an embodiment, a ity of beads are
obtained, and at least 80%, e.g., at least 85%, 90%, 95 %, 98%, 99%, or 100%, of the plurality
contains no more than one cell per droplet. In an embodiment, a plurality of beads are obtained, and
at least 80%, e.g., at least 85%, 90%, 95%, 98%, 99%, or 100%, of the plurality contains at least one
bead per droplet. Typically, the occupancy of drops is no more than one cell per droplet, and at least
one bead per droplet.
Cell Lysis
After encapsulation, the droplets can be ted to facilitate cell lysis. In an embodiment,
an emulsion (e. g., containing coalesce different on phases) is heated to ed lysis efficiency
in the presence of a detergent (e.g., Tween20). In an embodiment, the emulsion is incubated at a
temperature between 40°C and 80°C, e. g., between 40°C and 60°C, 50°C and 70°C, or 60°C and
80°C, e. g., at 40°C, 50°C, 60°C, 70°C, or 80°C. In an embodiment, the emulsion is incubated for 5 to
60 minutes, e.g., 10 to 45 minutes, 15 to 30 s, 5 to 30 minutes, or 30 to 50 minutes. In an
embodiment, the cell is lysed by heat. In an embodiment, the cell is lysed by an enzyme. Typically,
after the cell is lysed, mRNA is ed and is captured on a bead by annealing to the
oligonucleotides on the bead.
Bead Recovery
Emulsions (e. g., ning coalesce different solution ) can be broken using a drop
destabilizing reagent, e.g., perfluorooctanol (PFO). In an embodiment, the bead-containing aqueous
phase is recovered. In an embodiment, the bead is a magnetic bead, and is isolated using magnet. In
an embodiment, the bead is washed and resuspended in a buffer (e. g., Tris, pH 7.5). In an
embodiment, the bead is kept cold to reduce dissociation of mRNA from the bead.
Reverse Transcription
Reverse transcription can be performed using standard methods. In an embodiment, the
reverse transcription is performed in a non-emulsion reaction. In an embodiment, the reverse
transcription is performed in an emulsion reaction. In a typical embodiment, the reverse transcription
step is performed within the PCR drop. For example, mRNA-beads are ulated into drops with
both reverse transcriptase and DNA polymerase to facilitate cDNA formation and dsDNA
amplification. In an embodiment, the bead with captured mRNA is resuspended in a buffer-enzyme
mix (e. g., Superscript II RT) and incubated at 35°C to 45°C (e. g., at 40°C) for 10 to 60 minutes (e.g.,
minutes) to tate reverse transcription. In an ment, the oligonucleotide coupled to the
bead is used as a primer for the synthesis of the first strand cDNA. In an embodiment, the bead is
washed with a buffer (e.g., Tris, pH 7.5) after reverse transcription.
Bead Encapsulationfor PCR
Beads can be encapsulated individually into droplets. In an embodiment, the volume of the
droplet is from 5 pL to 500 pL, e. g., from 5 pL to 400 pL, from 5 pL to 300 pL, from 5 pL to 200 pL,
from 5 pL to 100 pL, from 5 pL to 50 pL, from 5 pL to 25 pL, from 400 pL to 500 pL, from 300 pL to
500 pL, from 200 pL to 500 pL, from 100 pL to 500 pL, from 50 pL to 500 pL, from 25 pL to 500 pL,
from 10 pL to 500 pL, from 10 pL to 400 pL, from 25 pL to 300 pL, from 50 pL to 200 pL, or from
10 pL to 50 pL. In an embodiment, the volume of the droplet is from 10 pL to 50 pL.
In an embodiment, the droplet is a water-in-oil droplet. In an embodiment, the droplet is
present in a carrier (e.g, oil) phase, e.g., a carrier phase comprising 3MTM HFE-7500 with about 1%
fluorosurfactant (RAN Biotechnologies).
In an embodiment, the droplet contains one bead after encapsulation. In an embodiment, a
plurality of beads are obtained, and at least 80%, e.g., at least 85%, 90%, 95%, 98%, 99%, or 100%,
of the plurality contains no more than one bead per droplet.
PCR Reaction
In an embodiment, a PCR on is performed. In an embodiment, the PCR reaction is
performed in a droplet comprising a bead that is coupled with cDNA, a DNA polymerase,
oligonucleotides (e.g., for amplification of the cDNA), and a buffer.
Exemplary DNA polymerases that can be used in the on include, but are not limited to,
n® High-Fidelity DNA rase (NEB), Q5® High-Fidelity DNA Polymerase (NEB), Pfu
DNA polymerase, KAPA DNA polymerase, Vent® DNA polymerase, or Taq DNA rase.
In an embodiment, the PCR product contains an scFv cassette. In an embodiment, the scFv
cassette is ucted as VL-Linker-VH. Without wishing to be bound by theory, it is believed that
in an embodiment, the order can be switched to VH-Linker-VL, with no cant impact on
expression or function. In an embodiment, the scFv cassette is constructed as VH-Linker-VL.
Similarly, the PCR product can contain a cassette constructed as or chain-Linker-B chain or [3
chain-Linker-a chain, or y chain-Linker-8 chain or 6 chain-Linker-y chain.
In an ment, a primer for a target variable region sequence described herein can contain
(e.g., from 5’ to 3’): a first sequence that is complementary to the sequence of an oligonucleotide
attached to a e substrate, a spacer (e.g., a spacer described herein, e.g., a PEG spacer), a
sequence that is complementary to at least a portion of the first sequence, a sal priming
sequence, and a sequence complementary to the target le region sequence.
In an embodiment, the reverse primer for the VL ce contains one, two, or all of the
ing: (a) an overhang sequence encoding a linker sequence (b) at least one modified nucleotide
2017/068204
(e. 3., 3 consecutive nucleotides with 2’-O-methyl modification), e. g., in the overhang; or (c) a 5’-
phosphate. In an ment, the d primer for the VH ce contains one, two, or all of
the following: (a) an overhang sequence encoding a linker sequence (b) at least one modified
nucleotide (e.g., 3 consecutive nucleotides with 2’-O-methyl modification), e. g., in the overhang; or
(c) a 5’-phosphate.
In an embodiment, the reverse primer for the VH sequence contains one, two, or all of the
following: (a) an overhang sequence encoding a linker sequence (b) at least one modified nucleotide
(e. g., 3 consecutive nucleotides with 2’-O-methyl modification), e. g., in the overhang; or (c) a 5’-
phosphate. In an embodiment, the forward primer for the VL sequence contains one, two, or all of the
following: (a) an overhang sequence encoding a linker sequence (b) at least one modified nucleotide
(e.g., 3 consecutive nucleotides with 2’-O-methyl modification), e. g., in the overhang; or (c) a 5’-
ate.
Similarly, in an embodiment, the reverse primer for the Qt chain (or 7 chain) sequence contains
one, two, or all of the following: (a) an overhang sequence encoding a linker sequence (b) at least one
modified nucleotide (e.g., 3 consecutive nucleotides with 2’-O-methyl modification), e. g., in the
overhang; or (c) a 5’-phosphate. In an embodiment, the d primer for the B chain (or 5 chain)
sequence contains one, two, or all of the following: (a) an overhang sequence encoding a linker
sequence (b) at least one modified nucleotide (e. g., 3 consecutive nucleotides with 2’-O-methyl
modification), e. g., in the ng; or (c) a 5’-phosphate.
rly, in an embodiment, the reverse primer for the B chain (or 5 chain) ce contains
one, two, or all of the following: (a) an overhang sequence ng a linker sequence (b) at least one
modified nucleotide (e.g., 3 consecutive tides with ethyl cation), e. g., in the
overhang; or (c) a 5’-phosphate. In an embodiment, the forward primer for the a chain (or 7 chain)
sequence contains one, two, or all of the following: (a) an overhang sequence encoding a linker
sequence (b) at least one modified nucleotide (e. g., 3 consecutive nucleotides with 2’-O-methyl
ation), e. g., in the overhang; or (c) a 5’-phosphate.
In an embodiment, the thermocycling is performed with emulsion (e. g., in a PCR tube). In an
embodiment, the thermocycling is performed using the following conditions: initial denaturation at
95-98°C for 30 seconds to 2 minutes; 10-30 cycles of: denaturation at 95-98°C for 10-30 seconds,
primer annealing at 50-60°C for 10-30 seconds, and polymerase ion at 72°C for 30 seconds. In
an embodiment, the reaction undergoes a slow cooling to facilitate capture of PCR products onto
beads. The reaction can be hold at 4°C.
Bead Recovery
Emulsions (e.g., containing ce different solution phases) can be broken using a drop
destabilizing reagent, e. g., perfluorooctanol (PFO). In an embodiment, the bead-containing aqueous
phase is recovered. In an embodiment, the bead is a magnetic bead, and is isolated using magnet. In
an embodiment, the bead is washed and resuspended in a buffer (e. g., Tris, pH 7.5).
Bead Encapsulationfor Ligase Cycling
Beads can be encapsulated individually into droplets. In an embodiment, the volume of the
droplet is from 5 pL to 500 pL, e. g., from 5 pL to 400 pL, from 5 pL to 300 pL, from 5 pL to 200 pL,
from 5 pL to 100 pL, from 5 pL to 50 pL, from 5 pL to 25 pL, from 400 pL to 500 pL, from 300 pL to
500 pL, from 200 pL to 500 pL, from 100 pL to 500 pL, from 50 pL to 500 pL, from 25 pL to 500 pL,
from 10 pL to 500 pL, from 10 pL to 400 pL, from 25 pL to 300 pL, from 50 pL to 200 pL, or from
10 pL to 50 pL. In an embodiment, the volume of the droplet is from 10 pL to 50 pL.
In an embodiment, the droplet is a water-in-oil droplet. In an embodiment, the droplet is
present in a carrier (e.g., oil) phase, e. g., a carrier phase comprising 3MTM HFE-7500 with about 1%
fluorosurfactant (RAN Biotechnologies).
In an embodiment, the droplet contains one bead after encapsulation. In an embodiment, a
plurality of beads are ed, and at least 80%, e.g., at least 85%, 90%, 95%, 98%, 99%, or 100%,
of the plurality contains no more than one bead per t.
Ligase Cycling Reaction
In an embodiment, a ligase cycling reaction is med. In an embodiment, the ligase
cycling reaction is med in a droplet comprising a bead that is coupled with PCR product, a
Splint oligonucleotide (e. g., complementary and anneals to 3’ terminus of one strand (e. g., “top” VL
strand) and 5’ us of another strand (e. g., “top” VH strand)), a thermostable ligase, and one or
more reaction components that supports ligase enzymatic activity (e. g., NAD).
Exemplary ligases (e. g., thermostable ligases) that can be used in the reaction e, but are
not d to, Taq DNA ligase, Pfu DNA ligase, Ampligase® thermostable DNA , Tsc DNA
ligase, Rma DNA ligase, Tfi DNA ligase, or Tth DNA .
In an embodiment, the cycling is performed with emulsion (e. g., in a PCR tube). In an
embodiment, the thermocycling is performed using the following conditions: 3- 15 cycles of:
denaturation at 90-95°C for 30 seconds, and annealing and ligation at 50-60°C for 1-3 s. The
reaction can be hold at 4°C.
Recovery ofAqueous Portion
Emulsions (e.g., containing coalesce different solution phases) can be broken using a drop
destabilizing reagent, e. g., perfluorooctanol (PFO). In an embodiment, the aqueous portion (e. g.,
ning linked product, and optionally, non-linked product) is recovered. In an embodiment, the
bead is discarded.
Purification ofLinked Product
Linked product (e.g., representing ly linked VL—linker-VH) can be purified from non-
linked products (e.g., non-linked VH and VL). The ligated products are ted from non-ligated
products by size separation. For example, ring PAGE (polyacrylamide gel electrophoresis),
denaturing HPLC-SEC, agarose gel electrophoresis or AMPure XP beads can be used. The linked
product 900 bp) is isolated from non-linked product (~350-500 bp). For denaturing PAGE
purification, the ligated band is cut out from the gel and an elecro-elution is performed to extract
DNA from gel slice (Bio-Rad Electro-Elutor).
Amplification ofPurified Linked Product
The purified linked product can be amplified, e.g., by PCR. For example, the purified linked
product is amplified by PCR using a DNA polymerase (e.g., Taq polymerase) under standard
conditions with oligonucleotides that anneal the outer termini of the ligated product.
The final PCR product can be introduced to yeast or ian cells using standard methods
(e.g., electroporation with expression vector) to create a natively paired library derived from
biological sources.
Exemplary steps in a method of making a nucleic acid sing a sequence that encodes a
heavy chain element (HC element) of an antibody heavy chain variable region (HCVR) and a light
chain element (LC element) of an antibody light chain variable region , and wherein the
HCVR and LCVR are matched, are illustrated in FIGS. 2A-2D.
Additional Exemplary Methods
In an aspect, the disclosure features a method of making a nucleic acid ce comprising a
sequence that encodes a heavy chain element (HC t) of an dy heavy chain variable
region (HCVR) and a light chain element (LC element) of an antibody light chain variable region
(LCVR), and n the HCVR and LCVR are matched, the method sing carrying out the
following steps from A1, B1, C1, and D1, thereby making a nucleic acid ce comprising
a sequence that encodes an HC element of an HCVR and a LC element of an LCVR, wherein the
HCVR and LCVR are d.
In an aspect, the disclosure features a method of making a nucleic acid sequence comprising a
sequence that encodes a heavy chain element (HC element) of an antibody heavy chain variable
region (HCVR) and a light chain element (LC element) of an antibody light chain variable region
(LCVR), and wherein the HCVR and LCVR are matched, the method comprising carrying out the
following steps from A1, B1, C1, D2, and E1, thereby making a nucleic acid sequence
comprising a sequence that encodes an HC element of an HCVR and a LC t of an LCVR,
wherein the HCVR and LCVR are matched.
In an , the disclosure features a method of making a nucleic acid sequence comprising a
sequence that encodes a heavy chain element (HC element) of an dy heavy chain variable
region (HCVR) and a light chain element (LC element) of an antibody light chain variable region
(LCVR), and wherein the HCVR and LCVR are matched, the method comprising carrying out the
following steps from A1, B1, C2, and D3, y making a nucleic acid sequence comprising
a sequence that encodes an HC element of an HCVR and a LC element of an LCVR, wherein the
HCVR and LCVR are matched.
In an aspect, the disclosure features a method of making a nucleic acid sequence comprising a
ce that encodes a heavy chain element (HC element) of an antibody heavy chain variable
region (HCVR) and a light chain element (LC element) of an antibody light chain variable region
, and wherein the HCVR and LCVR are matched, the method comprising carrying out the
following steps from A1, B1, C2, D4 and E2, thereby making a nucleic acid ce
comprising a ce that encodes an HC element of an HCVR and a LC element of an LCVR,
wherein the HCVR and LCVR are matched.
In an aspect, the disclosure features a method of making a nucleic acid sequence comprising a
sequence that encodes a heavy chain t (HC element) of an antibody heavy chain variable
region (HCVR) and a light chain t (LC element) of an antibody light chain variable region
(LCVR), and wherein the HCVR and LCVR are matched, the method comprising carrying out the
following steps from A1, B1, C3, and D5, thereby making a nucleic acid sequence comprising
a ce that encodes an HC t of an HCVR and a LC element of an LCVR, wherein the
HCVR and LCVR are d.
In an aspect, the sure features a method of making a nucleic acid sequence comprising a
sequence that encodes a heavy chain element (HC element) of an antibody heavy chain variable
region (HCVR) and a light chain element (LC element) of an antibody light chain variable region
(LCVR), and wherein the HCVR and LCVR are matched, the method comprising carrying out the
following steps from A1, B1, C3, D6 and E3, thereby making a nucleic acid sequence
comprising a sequence that encodes an HC element of an HCVR and a LC element of an LCVR,
wherein the HCVR and LCVR are matched.
In the aforesaid exemplary methods, the cDNA is typically not ed on the substrate (e.g.,
bead) in this workflow concept. For example, mRNA dissociates from the substrate (e.g., bead), then
cDNA is made in solution in the isolated on site (e.g., micro-chamber), e.g., in the drop, and
then PCR product is made from cDNA as template in solution in the isolated reaction site (e.g., micro-
chamber), e. g., in drop. In an embodiment, the method includes an RT-PCR reaction, where both
enzymatic steps occur in solution in drop. In the aforesaid exemplary methods, the amplified products
are typically captured onto the substrate (e.g., bead), which can facilitate transitioning the paired
products into the next isolated reaction site (e.g., micro-chamber), e.g., the next drop.
Antibody Molecules
Disclosed herein are antibody molecules and libraries of antibody molecules. In an
embodiment, the antibody molecule or library of antibody molecules are made by a method bed
herein.
As used herein, the term “antibody le” refers to a protein, e.g., an immunoglobulin
chain or a fragment thereof, comprising at least one imrnunoglobulin variable domain sequence. The
term “antibody molecule” includes, for example, full-length, mature antibodies and antigen-binding
fragments of an antibody. For example, an antibody le can include a heavy (H) chain variable
domain sequence (abbreviated herein as VH), and a light (L) chain variable domain sequence
(abbreviated herein as VL). In another example, an dy molecule includes two heavy (H) chain
variable domain sequences and two light (L) chain variable domain ce, thereby forming two
antigen binding sites, such as Fab, Fab’, F(ab’)2, Fc, Fd, Fd’, Fv, single chain antibodies (scFv for
example), single variable domain antibodies, diabodies (Dab) ent and bispecific), and chimeric
(e.g., zed) antibodies, which may be produced by the modification of whole antibodies or
those synthesized de novo using recombinant DNA technologies. These functional antibody
nts retain the ability to selectively bind with their respective n or receptor. Antibodies
and dy fragments can be from any class of antibodies including, but not limited to, IgG, IgA,
IgM, IgD, and IgE, and from any ss (e.g., IgGl, IgGZ, IgG3, and IgG4) of antibodies. The
antibody molecules can be monoclonal or polyclonal. The antibody le can also be a human,
humanized, CDR-grafted, or in vitro ted antibody. The antibody molecule can have a heavy
chain constant region chosen from, e.g., IgGl, IgGZ, IgG3, or IgG4. The antibody molecule can also
have a light chain chosen from, e.g., kappa or lambda. The term “immunoglobulin” (lg) is used
interchangeably with the term “antibody” herein.
Examples of antigen-binding fragments include: (i) a Fab fragment, a monovalent fragment
consisting of the VL, VH, CL and CH1 domains; (ii) a F(ab')2 nt, a bivalent fragment
comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment
consisting of the VH and CH1 domains; (iv) a Fv fragment consisting of the VL and VH domains of a
single arm of an antibody, (v) a diabody (dAb) fragment, which consists of a VH domain; (vi) a
camelid or camelized variable domain; (vii) a single chain Fv , see e.g., Bird er al. (1988)
Science 242:423-426; and Huston er al. (1988) Proc. Natl. Acad. Sci. USA 9-5883); (viii) a
single domain antibody. These antibody fragments may be obtained using any suitable method,
including several conventional techniques known to those with skill in the art, and the fragments can
be screened for utility in the same manner as are intact antibodies.
The term “antibody” includes intact molecules as well as functional fragments thereof.
Constant regions of the dies can be altered, e.g., mutated, to modify the properties of the
antibody (e.3., to increase or decrease one or more of: Fc receptor binding, antibody ylation,
the number of cysteine residues, effector cell function, or complement function).
The antibody molecule can be a single chain antibody. A single-chain dy (scFV) may
be engineered (see, for example, Colcher, D. et al. (1999) Ann N Y Acad Sci 880:263-80; and Reiter,
Y. (1996) Clin Cancer Res 2:245-52). The single chain antibody can be dimerized or multimerized to
generate multivalent antibodies having specificities for ent epitopes of the same target protein.
The antibody molecules disclosed herein can also be single domain dies. Single
domain antibodies can include antibodies whose mentary determining regions are part of a
single domain polypeptide. Examples include, but are not limited to, heavy chain dies,
antibodies naturally devoid of light chains, single domain dies derived from tional 4-
chain antibodies, engineered antibodies and single domain scaffolds other than those derived from
antibodies. Single domain antibodies may be any of the art, or any future single domain antibodies.
Single domain antibodies may be derived from any species including, but not limited to mouse,
human, camel, llama, fish, shark, goat, rabbit, and bovine. According to some aspects, a single
domain antibody is a naturally occurring single domain antibody known as heavy chain antibody
devoid of light chains. Such single domain antibodies are disclosed in WO 94/04678, for example.
For clarity reasons, this variable domain d from a heavy chain antibody naturally devoid of light
chain is known herein as a VHH or nanobody to distinguish it from the conventional VH of four chain
immunoglobulins. Such a VHH molecule can be d from antibodies raised in Camelidae
species, for example in camel, llama, dromedary, alpaca and guanaco. Other species besides
Camelidae may e heavy chain antibodies naturally devoid of light chain; such VHHs are also
contemplated.
The VH and VL regions can be subdivided into regions of hypervariability, termed
“complementarity determining regions” (CDR), interspersed with s that are more conserved,
termed work regions” (FR or FW). The terms “complementarity determining region,” and
“CDR,” as used herein refer to the sequences of amino acids within antibody variable regions which
confer antigen specificity and binding affinity. As used herein, the terms “framework,” “FW” and
“FR” are used interchangeably.
The extent of the framework region and CDRs has been precisely defined by a number of
s (see, Kabat, E. A., et al. (1991) Sequences of Proteins of Immunological Interest, Fifth
Edition, US. Department of Health and Human Services, NIH Publication No. 2; Chothia, C.
et al. (1987) J. Mol. Biol. 196:901-917; and the AbM ion used by Oxford Molecular’s AbM
antibody modeling software. See, generally, e.g., Protein Sequence and Structure Analysis of
Antibody Variable Domains. In: Antibody Engineering Lab Manual (Ed.: Duebel, S. and
Kontermann, R., Springer-Verlag, Heidelberg). In an embodiment, the following definitions are used:
AbM definition of CDRl of the heavy chain variable domain and Kabat definitions for the other
CDRs. In an embodiment, Kabat definitions are used for all CDRs. In addition, embodiments
described with respect to Kabat or AbM CDRs may also be ented using Chothia hypervariable
loops. Each VH and VL typically includes three CDRs and four FRs, arranged from amino-terminus
to carboxy-terminus in the following order: FRl, CDRl, FR2, CDRZ, FR3, CDR3, and FR4.
As used herein, an “immunoglobulin variable domain sequence” refers to an amino acid
sequence which can form the structure of an immunoglobulin variable domain. For example, the
sequence may include all or part of the amino acid sequence of a naturally-occurring variable domain.
For example, the sequence may or may not include one, two, or more N- or C-terminal amino acids,
or may e other tions that are compatible with formation of the protein structure.
The term “antigen-binding region” refers to the part of an antibody molecule that comprises
determinants that form an interface that binds to an antigen, or an epitope thereof. With respect to
proteins (or protein mimetics), the antigen-binding region typically includes one or more loops (of at
least, e.g., four amino acids or amino acid mimics) that form an interface that binds to the antigen.
Typically, the antigen-binding region of an antibody molecule includes at least one or two CDRs
andfor hypervariable loops, or more typically at least three, four, five or six CDRs and/or
hypervariable loops.
The terms te” or “cross-compete” are used interchangeably herein to refer to the
ability of an antibody molecule to interfere with binding of another antibody molecule, to a target.
The interference with binding can be direct or indirect (e. g., h an allosteric modulation of the
dy molecule or the target). The extent to which an antibody molecule is able to ere with
the binding of another dy molecule to the target, and therefore whether it can be said to
compete, can be determined using a ition binding assay, for example, a FACS assay, an ELISA
or BIACORE assay. In an embodiment, a competition binding assay is a quantitative ition
assay. In an embodiment, a first antibody molecule is said to compete for binding to the target with a
second dy molecule when the binding of the first antibody molecule to the target is reduced by
% or more, e.g., 20% or more, 30% or more, 40% or more, 50% or more, 55% or more, 60% or
more, 65% or more, 70% or more, 75% or more, 80% or more, 85% or more, 90% or more, 95% or
more, 98% or more, 99% or more in a competition binding assay (e.g., a competition assay described
herein).
The terms “monoclonal antibody” or “monoclonal antibody composition” as used herein refer
to a preparation of antibody les of single molecular composition. A monoclonal antibody
composition ys a single binding city and affinity for a particular epitope. A monoclonal
antibody can be made by hybridoma logy or by s that do not use hybridoma technology
(e.g., recombinant methods).
An “effectively human” protein is a protein that does not evoke a neutralizing dy
response, e.g., the human anti-murine antibody (HAMA) response. HAMA can be problematic in a
number of circumstances, e. g., if the antibody molecule is administered repeatedly, e. g., in treatment
of a chronic or recurrent disease condition. A HAMA response can make repeated antibody
administration potentially ineffective because of an increased antibody clearance from the serum (see,
e.g., Saleh et al., Cancer Immunol. Immunother. 32:180-190 (1990)) and also because of ial
allergic reactions (see, e. g., LoBuglio et at, Hybridoma, 5:5117-5123 (1986)).
The antibody molecule can be a polyclonal or a monoclonal antibody. In some embodiments,
the antibody can be recombinantly produced, e. g., produced by any suitable phage display or
combinatorial methods.
Various phage display and combinatorial methods for generating antibodies are known in the
art (as described in, e. g., Ladner et al. U.S. Patent No. 5,223,409; Kang et al. International ation
No. W0 92/18619; Dower et al. International Publication No. WO 91/17271; Winter et al.
International ation WO 92/20791; Markland et al. ational Publication No. W0 92/ 15679;
ing et al. International ation WO 93/01288; McCafferty et al. International Publication
No. WO 92/01047; Garrard et al. International Publication No. WO 92I09690; Ladner et al.
International Publication No. WO 09; Fuchs et al. (1991) Bio/Technology 9: 1370-1372; Hay et
al. (1992) Hum Antiboa’ omas 3:81-85; Huse et al. (1989) e 246: 281; Griffths er
al. (1993) EMBO J 12:725-734; Hawkins et al. (1992) JMol Biol 226:889-896; Clackson et al. (1991)
Nature 352:624-628; Gram et al. (1992) PNAS 89:3576-3580; Garrad et al. (1991) Bio/Technology
9: 1373-1377; Hoogenboom et al. (1991) Nuc Acid Res 19:4133-4137; and Barbas et al. (1991) PNAS
88:7978-7982, the contents of all of which are incorporated by reference herein).
In an embodiment, the antibody molecule is a fully human antibody (e. g., an antibody made
in a mouse which has been genetically engineered to produce an antibody from a human
immunoglobulin sequence), or a non-human antibody, e. g., a rodent (mouse or rat), goat, primate
(e. g., monkey), camel dy. In an embodiment, the non-human antibody is a rodent (mouse or rat
antibody). Methods of producing rodent antibodies are known in the art.
Human monoclonal antibodies can be generated using transgenic mice carrying the human
immunoglobulin genes rather than the mouse system. Splenocytes from these transgenic mice
immunized with the n of interest are used to produce hybridomas that secrete human mAbs with
specific affinities for epitopes from a human protein (see e. g., Wood et al. International Application
WO 91/00906, Kucherlapati et al. PCT ation W0 91/ 10741; g et al. International
Application WO 92/03918; Kay et al. International Application 92/03917; Lonberg, N. et al. 1994
Nature 368:856-859; Green, L.L. et al. 1994 Nature Genet. 7:13-21; Morrison, S.L. et al. 1994 Proc.
Natl. Acad. Sci. USA 81:6851-6855; Bruggeman et al. 1993 Year Immunol 7:33-40; Tuaillon et al.
1993 PNAS 90:3720—3724; Bruggeman et al. 1991 Eur J Immunol 3-1326).
An antibody can be one in which the variable , or a portion thereof, e. g., the CDRs, are
generated in a non-human sm, e.g., a rat or mouse. ic, CDR-grafted, and zed
antibodies are within the invention. Antibodies generated in a non-human organism, e. g., a rat or
2017/068204
mouse, and then ed, e.g., in the variable framework or constant region, to decrease antigenicity
in a human are within the invention.
Chimeric antibodies can be produced by any suitable recombinant DNA technique. Several
are known in the art (see Robinson et al., International Patent Publication PCT/US86/02269; Akira, et
al., European Patent Application 7; chi, M., European Patent Application 171,496;
Morrison et al., European Patent Application 173,494; Neuberger et al., International Application WO
86301533; Cabilly et al. US. Patent No. 4,816,567; Cabilly et al., European Patent Application
125,023; Better et al. (1988 Science 240: 1041-1043); Liu et al. (1987) PNAS 84:3439-3443; Liu et
al., 1987, J. l. 139:3521-3526; Sun et al. (1987) PNAS -218; Nishimura et al., 1987,
Canc. Res. 47:999-1005; Wood et al. (1985) Nature 314:446—449; and Shaw et al., 1988, J. Natl
Cancer Inst. 80: 1553- 1559).
A humanized or CDR-grafted antibody will have at least one or two but generally all three
recipient CDRs (of heavy and or light immunoglobulin chains) replaced with a donor CDR. The
antibody may be replaced with at least a n of a non-human CDR or only some of the CDRs may
be replaced with man CDRs. It is only necessary to replace the number of CDRs required for
binding of the humanized antibody to an n. In an embodiment, the donor will be a rodent
antibody, e. g., a rat or mouse antibody, and the recipient will be a human ork or a human
sus framework. Typically, the immunoglobulin providing the CDRs is called the “donor” and
the immunoglobulin providing the framework is called the “acceptor.” In some embodiments, the
donor immunoglobulin is a non-human (e.g., rodent). The acceptor ork is typically a
naturally-occurring (e.g., a human) framework or a consensus framework, or a sequence about 85% or
higher, e.g., 90%, 95%, 99% or higher identical thereto.
As used herein, the term “consensus sequence” refers to the sequence formed from the most
frequently occurring amino acids (or nucleotides) in a family of related sequences (See e.g., er,
From Genes to Clones (Verlagsgesellschaft, Weinheim, y 1987). In a family of proteins, each
position in the consensus sequence is occupied by the amino acid occurring most frequently at that
on in the family. If two amino acids occur equally frequently, either can be included in the
consensus sequence. A “consensus framework” refers to the framework region in the consensus
immunoglobulin sequence.
An antibody can be humanized by any suitable method, and several such methods known in
the art (see e.g., Morrison, S. L., 1985, Science 229: 1202-1207, by Oi et al., 1986, BioTechniqaes
4:214, and by Queen et al. US 5,585,089, US 5,693,761 and US 5,693,762, the contents of all of
which are hereby incorporated by reference).
zed or CDR-grafted antibodies can be produced by CDR-grafting or CDR
substitution, wherein one, two, or all CDRs of an immunoglobulin chain can be replaced. See e.g.,
US. Patent 5,225,539; Jones et al. 1986 Nature 321 :552-525; yan et al. 1988 Science
239:1534; Beidler et al. 1988 J. Immunol. 141:4053-4060; Winter US 5,225,539, the contents of all of
which are hereby expressly incorporated by reference. Winter describes a CDR-grafting method
which may be used to prepare humanized antibodies (UK Patent ation GB 2188638A, filed on
March 26, 1987; Winter US 5,225,539), the contents of which is expressly incorporated by reference.
Also provided are humanized antibodies in which specific amino acids have been substituted,
deleted or added. Criteria for selecting amino acids from the donor are described in, e. g., US
,585,089, e.g., columns 12-16 of US 5,585,089, the contents of which are hereby orated by
reference. Other techniques for zing antibodies are described in Padlan et al. EP 519596 A1,
published on December 23, 1992.
In an embodiment, the antibody molecule has a heavy chain constant region chosen from,
e.g., the heavy chain constant regions of IgG1, IgG2 (e.g., IgG2a), IgG3, IgG4, IgM, IgAl, IgA2,
IgD, and IgE; particularly, chosen from, e.g., the (e.g., human) heavy chain nt regions of IgG1,
IgG2, IgG3, and IgG4. In another embodiment, the antibody molecule has a light chain constant
region chosen from, e. g., the (e.g., human) light chain nt regions of kappa or lambda. The
constant region can be d, e. g., mutated, to modify the properties of the dy molecule (e.g.,
to increase or decrease one or more of: Fc receptor binding, antibody glycosylation, the number of
cysteine residues, effector cell on, and/or ment function). In an embodiment, the
antibody molecule has effector function and can fix complement. In another embodiment, the
antibody molecule does not recruit effector cells or fix complement. In certain embodiments, the
antibody le has reduced or no ability to bind an Fc receptor. For example, it may be an isotype
or subtype, fragment or other mutant, which does not support binding to an Fc receptor, e. g., it has a
mutagenized or deleted Fc receptor binding region.
In an embodiment, a constant region of the antibody molecule is altered. Methods for altering
an antibody constant region are known in the art. Antibody molecules with altered function, e.g.
altered affinity for an effector ligand, such as FcR on a cell, or the C1 component of complement can
be produced by replacing at least one amino acid residue in the constant portion of the antibody with a
different residue (see e.g., EP 388,151 A1, US. Pat. No. 5,624,821 and US. Pat. No. 5,648,260, the
ts of all of which are hereby orated by reference). Amino acid mutations which stabilize
antibody ure, such as S228P (EU nomenclature, S241P in Kabat nomenclature) in human IgG4
are also contemplated. Similar type of alterations could be described which if applied to the murine,
or other species immunoglobulin would reduce or eliminate these functions.
In an ment, the only amino acids in the antibody molecule are canonical amino acids.
In an embodiment, the antibody molecule comprises naturally-occurring amino acids; s,
derivatives and congeners thereof; amino acid analogs having variant side ; and/or all
stereoisomers of any of any of the foregoing. The antibody molecule may se the D- or L-
optical isomers of amino acids and peptidomimetics.
A polypeptide of an dy molecule described herein may be linear or branched, it may
comprise modified amino acids, and it may be interrupted by non-amino acids. The antibody
WO 19402
molecule may also be modified; for example, by disulfide bond formation, glycosylation, lipidation,
acetylation, phosphorylation, or any other manipulation, such as ation with a labeling
component. The polypeptide can be isolated from natural s, can be a produced by recombinant
techniques from a eukaryotic or prokaryotic host, or can be a product of synthetic procedures.
The antibody molecule described herein can be used alone in unconjugated form, or can be
bound to a substance, e. g., a toxin or moiety (e.g., a eutic drug; a compound emitting radiation;
molecules of plant, fungal, or bacterial origin; or a biological protein (e.g., a protein toxin) or particle
(e.g., a recombinant Viral particle, e.g., Via a Viral coat n). For example, the antibody molecule
can be coupled to a radioactive isotope such as an (1-, [3-, or y-emitter, or a B-and y-emitter.
An antibody molecule can be derivatized or linked to r functional molecule (e.g.,
another peptide or protein). As used herein, a “derivatized” antibody le is one that has been
modified. Methods of derivatization include but are not limited to the addition of a fluorescent
moiety, a radionucleotide, a toxin, an enzyme or an affinity ligand such as biotin. Accordingly, the
antibody molecules are intended to include derivatized and otherwise modified forms of the
antibodies described herein, including immunoadhesion molecules. For example, an antibody
molecule can be functionally linked (by chemical ng, genetic fusion, noncovalent association or
otherwise) to one or more other molecular entities, such as another antibody (e.g., a bispecific
antibody or a diabody), a detectable agent, a toxin, a pharmaceutical agent, andior a protein or peptide
that can mediate association of the antibody or antibody portion with another molecule (such as a
streptavidin core region or a polyhistidine tag).
Some types of derivatized antibody molecule are produced by crosslinking two or more
antibodies (of the same type or of different types, e.g., to create bispecific antibodies). le
crosslinkers include those that are heterobifunctional, having two distinctly ve groups separated
by an appropriate spacer (e.g., m-maleimidobenzoyl-N-hydroxysuccinimide ester) or
homobifunctional (e.g., disuccinimidyl te). Such s are available from Pierce Chemical
Company, rd, Ill.
Useful able agents with which an antibody molecule may be tized (or labeled) to
include fluorescent compounds, various enzymes, prosthetic groups, luminescent als,
bioluminescent materials, fluorescent emitting metal atoms, e.g., europium (Eu), and other anthanides,
and radioactive materials (described below). Exemplary fluorescent detectable agents include
fluorescein, fluorescein isothiocyanate, rhodamine, 5dimethylamine-l-napthalenesulfonyl chloride,
phycoerythrin and the like. An antibody may also be derivatized with able enzymes, such as
alkaline atase, horseradish peroxidase, B-galactosidase, cholinesterase, glucose oxidase
and the like. When an antibody is derivatized with a detectable enzyme, it is detected by adding
additional reagents that the enzyme uses to produce a detectable reaction product. For e, when
the able agent horseradish peroxidase is present, the addition of hydrogen peroxide and
diaminobenzidine leads to a colored reaction product, which is detectable. An antibody molecule may
also be derivatized with a prosthetic group (e.g., avidin/biotin and avidin/biotin). For e,
an antibody may be derivatized with biotin, and ed through indirect measurement of avidin or
streptavidin binding. Examples of suitable fluorescent materials include umbelliferone, fluorescein,
fluorescein ocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or
phycoerythrin; an example of a luminescent al includes luminol; and examples of
bioluminescent materials include luciferase, luciferin, and aequorin.
Labeled antibody molecules can be used, for example, diagnostically and/or experimentally in
a number of contexts, including (i) to isolate a ermined antigen by standard techniques, such as
affinity chromatography or immunoprecipitation; (ii) to detect a predetermined antigen (e.g., in a
ar lysate or cell supernatant) in order to evaluate the abundance and pattern of expression of the
protein; (iii) to monitor protein levels in tissue as part of a al testing ure, e.g., to determine
the efficacy of a given treatment regimen.
An antibody molecule may be conjugated to another molecular entity, typically a label or a
therapeutic (e.g., antimicrobial (e. g., antibacterial or bactericidal), immunomodulatory,
immunostimularoty, cytotoxic, or cytostatic) agent or moiety. Radioactive es can be used in
diagnostic or therapeutic applications. Radioactive isotopes that can be coupled to the antibody
molecules include, but are not limited to or-, [3-, or y—emitters, or B-and y—emitters. Such radioactive
es include, but are not limited to iodine (1311 or 125I), m (90Y), lutetium (177Lu), actinium
(225Ac), praseodymium, astatine (leAt), rhenium (186Re), h (212Bi or 213Bi), indium (1 1 1In),
technetium (gngc), phosphorus (32F), rhodium (lgth), sulfur (35S) carbon (14C), tritium (3H),
chromium (5 lCr), chlorine (36Cl), cobalt (57Co or 58C0), iron (59Fe), selenium (75Se), or gallium (67Ga).
Radioisotopes useful as therapeutic agents include yttrium (90Y), lutetium (177Lu), actinium (225Ac),
praseodymium, astatine (leAt), rhenium (186Re), bismuth (212Bi or 213Bi), and rhodium (lgth).
sotopes useful as labels, e.g., for use in diagnostics, include iodine (1311 or 125I), indium (111111),
technetium (99mTc), phosphorus (32F), carbon (14C), and tritium (3H), or one or more of the therapeutic
es listed above.
The present disclosure es radiolabeled dy les and methods of labeling the
same. In an embodiment, a method of labeling an antibody le is disclosed. The method
includes contacting an antibody molecule, with a chelating agent, to thereby produce a conjugated
antibody. The conjugated antibody is radiolabeled with a radioisotope, e.g., 111Indium, 90Yttrium and
177Lutetium, to thereby produce a labeled antibody molecule.
In some aspects, this disclosure provides a method of making an antibody molecule disclosed
herein. The method includes: providing an n, or a fragment thereof; obtaining an antibody
molecule that specifically binds to the antigen; evaluating efficacy of the antibody molecule in
modulating activity of the n and/or organism expressing the antigen. The method can further
include administering the antibody molecule, including a derivative thereof (6.3., a humanized
antibody molecule) to a subject, e.g., a human.
This disclosure provides an isolated nucleic acid molecule encoding the above dy
molecule, vectors and host cells thereof. The nucleic acid le includes, but is not limited to,
RNA, genomic DNA and cDNA.
Other Binding Polypeptides
The disclosures herein are not ed to be limited to antibody molecules. The methods
described herein are broadly applicable to any binding polypeptides that have two or more chains
(e.g., having at least two paired or matched chains).
In an embodiment, the binding molecule comprises an X chain variable region and a Y chain
variable region. For example, in any of the s, embodiments, and tions herein, an antibody
heavy chain (or variable region) can be replaced with an X chain (or variable region), and an antibody
light chain (or variable region) can be replaced with a Y chain (or variable region).
In an aspect, the disclosure features a method of making a nucleic acid sequence comprising a
sequence that s an X chain element (XC element) of an antibody heavy chain variable region
(XCVR) and a Y chain element (YC t) of an antibody light chain variable region (YCVR), and
wherein the XCVR and YCVR are matched, the method comprising:
a) acquiring an isolated production reaction site, e.g., a production micro-chamber,
comprising:
i) an X chain (XC) strand, wherein the XC strand is a strand of an X chain double-stranded
cDNA (XC ds cDNA) comprising a segment that encodes an XC element of the XCVR from a cell,
e.g., an X chain variable region sequence (XCVRS); and
ii) a Y chain (YC) strand, wherein the YC strand is a strand of a Y chain -stranded
cDNA (YC ds cDNA) sing a t that encodes a YC element of the YCVR from the cell,
e.g., a Y chain variable region sequence (YCVRS), and
b) covalent linking, e.g., ligation, of an XC strand to a YC strand,
wherein the ed production reaction site, e.g., a production micro-chamber, does not
e a nucleic acid encoding a YCVR or an XCVR from a cell other than the cell (e.g., a different
cell),
thereby making a nucleic acid sequence comprising a sequence that encodes an XC element
of an XCVR and a YC element of a YCVR, n the XCVR and YCVR are matched.
“Matched,” as that term is used herein in connection with an X chain variable region and a Y
chain variable region, means they are from the same cell. In an embodiment, the X chain variable
region and the Y chain variable region can form a multimeric protein or a part of a multimeric n.
With respect to an element of an X chain variable region and an element of a Y chain variable region
it means that the X chain variable region and the Y chain variable region from which the elements are
derived are from the same cell.
An “X chain variable region sequence,” or “XCVRS,” as that term is used herein, refers to a
polypeptide comprising sufficient sequence to allow binding of another polypeptide (e.g., an antigen).
In embodiments the XCVRS can assemble with a Y chain variable region, and, e.g., bind antigen. A
“Y chain variable region sequence,” or “YCVRS,” as that term is used herein, refers to a polypeptide
comprising sufficient ce to allow binding of another polypeptide (e.g., an n). In
embodiments the YCVRS can assemble with an X chain variable region, and, e.g., bind antigen.
“Element” of an XC or YC le region, as that term is used herein, refers to a sequence
that encodes at least one amino acid. In an embodiment, an element comprises a CDR. In an
embodiment an element comprises a FW region.
In an embodiment, the XC element comprises, or consists of, an XCVRS. In an embodiment,
the YC element comprises, or consists of, a YCVRS.
In an embodiment, the XC ds cDNA comprises a segment that encodes an XCVRS. In an
embodiment, the YC ds cDNA comprises a segment that encodes a YCVRS. In an embodiment, the
XC ds cDNA ses a segment that encodes an XCVRS, and the YC ds cDNA comprises a
segment that encodes a YCVRS.
In an embodiment, the cell is an immune cell, e.g., a B cell, e.g., a human B cell. In an
embodiment, the cell is a ian cell or an avian cell.
In an embodiment, the nucleic acid ce is ured such that, when expressed, the XC
element and the YC t (e.g., the XCVRS and the YCVRS) form a functional antigen binding
molecule, e. g., a single chain or a complex of an XC and a YC. In an embodiment, the antigen
binding molecule is functional in vitro, ex vivo, or in vivo, e.g., as determined by a method or assay
bed herein.
In an embodiment, ing an isolated production reaction site, e.g., a production micro-
chamber, comprises:
a) acquiring a capture ate bound to: (i) a first double-stranded cDNA (ds cDNA)
comprising a strand that is complementary to a first mRNA that encodes an XCVR from a cell; and
(ii) a second ds cDNA comprising a strand complementary to a second mRNA encoding a YCVR
from the cell (the cDNA loaded capture substrate), and
b) maintaining the isolated production reaction site, e.g., the production micro-chamber,
under conditions that allow amplification of the first and second ds cDNAs, to e: a ity of
XC ds cDNAs comprising a segment that encodes an XC t of the XCVR from the cell, e.g., an
XCVRS; and a plurality of YC ds cDNAs comprising a segment that encodes a YC element of the
YCVR from the cell, e.g., a YCVRS.
In an embodiment, the XC ds cDNA is identical, or substantially identical, to the first ds
cDNA. For example, the sense strand of the XC ds cDNA is at least 80%, 85%, 90%, 95%, 98%,
99%, or 100% identical to, or differs by no more than 1, 2, 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50
nucleotides from, the sense strand of the first ds cDNA, and/or the antisense strand of the XC ds
cDNA is at least 80%, 85%, 90%, 95%, 98%, 99%, or 100% identical to, or differs by no more than 1,
2, 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50 nucleotides from, the antisense strand of the first ds cDNA.
In an embodiment, the YC ds cDNA is identical, or substantially identical, to the second ds
cDNA. For example, the sense strand of the YC ds cDNA is at least 80%, 85 %, 90%, 95%, 98%,
99%, or 100% identical to, or differs by no more than 1, 2, 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50
nucleotides from, the sense strand of the second ds cDNA, and/or the nse strand of the YC ds
cDNA is at least 80%, 85%, 90%, 95%, 98%, 99%, or 100% identical to, or differs by no more than 1,
2, 5, 10, 15, 20, 25, 30, 35, 40, 45 , or 50 nucleotides from, the antisense strand of the second ds
cDNA.
In an embodiment, the XC strand is a sense strand. In an embodiment, the YC strand is a
sense strand. In an embodiment, the XC strand is an antisense strand. In an embodiment, the YC
strand is an antisense . In an embodiment, both the XC strand and the YC strand are sense
strands. In an embodiment, both the XC strand and the YC strand are antisense strands.
In an embodiment, the capture substrate comprises a bead, e.g., a ic bead. In an
embodiment, the capture substrate comprises a moiety (e.g., an oligonucleotide) which binds to
cDNA, e.g., (i) a moiety which binds to the XC strand; (ii) a moiety which binds to the YC strand; or
(iii) both (i) and (ii). In an embodiment, the moiety which binds to the XC strand is different from the
moiety which binds to the YC strand, e.g., to tate creating conditions favorable to capturing
similar levels of each DNA molecule type. In an ment, the moiety which binds to the XC
strand is identical to the moiety which binds to the YC strand.
In an embodiment, the first mRNA and the second mRNA are disposed on an mRNA loaded
e substrate.
In an embodiment, the isolated production reaction site, e.g., the production micro-chamber,
comprises: a reagent mixture suitable for producing, from the first and second mRNAs (e.g., after the
first and second mRNAs are released from the mRNA loaded capture substrate into a solution), a first
cDNA comprising a segment that encodes an XC element of the XCVR of the cell, e.g., an XCVRS,
and a second cDNA comprising a segment that encodes a YC element of the YCVR of the cell, e.g., a
YCVRS.
In an embodiment, the ed production reaction site, e.g., production micro-chamber,
comprises s that mediate the production of the first ds cDNA. In an ment, the isolated
production reaction site, e.g., production micro-chamber, comprises primers that mediate the
tion of the second ds cDNA.
In an embodiment, a cDNA strand that is complementary to a first mRNA that encodes an
XCVR from a cell is made by reverse ription of the first mRNA. In an embodiment, a cDNA
strand that is complementary to a second mRNA that encodes a YCVR from a cell is made by reverse
transcription of the second mRNA.
In an embodiment, the reverse transcription takes place in the isolated production on
site, e.g., a production-micro chamber. In an embodiment, the reverse transcription takes place in an
isolated cell reaction site, e. g., a cell isolation micro-chamber. In an embodiment, the reverse
transcription takes place e the isolated production reaction site, e. g., a production micro-
chamber, or outside an isolated cell reaction site, e. g., a cell isolation micro-chamber. In an
embodiment, the reverse transcription takes place outside the isolated production reaction site, e.g., a
production-micro chamber, and outside an isolated cell reaction site, e.g., a cell isolation micro-
chamber. In an embodiment, the reverse transcription takes place outside an isolated reaction site,
e.g., outside a micro-chamber.
In an embodiment, the amplification ses 30 or fewer cycles, e.g., 20 or fewer cycles,
e.g., 15 or fewer, 14 or fewer, 13 or fewer, 12 or fewer, 11 or fewer, 10 or fewer, 9 or fewer, 8 or
fewer, 7 or fewer, 6 or fewer, or 5 or fewer cycles.
In an embodiment, the reverse transcription and/or amplification uses one or more primers,
e. g., comprising a sequence ic for an XCVRS and/or a YCVRS.
In an embodiment, the reverse transcription and/or amplification comprises using two or more
primers that e the production of the XC ds cDNA, wherein at least one primer comprises a
nucleotide modification, and wherein at least one primer does not se a nucleotide modification.
In an embodiment, the amplification comprises using two or more primers that mediate the production
of the YC ds cDNA, n at least one primer comprises a nucleotide modification, and wherein at
least one primer does not comprise a nucleotide modification.
In an embodiment, at least one primer ses a nucleotide modification, e.g., which
reduces, e. g., inhibits, DNA synthesis, e.g., by a DNA polymerase. In an embodiment, at least one
primer does not comprise a nucleotide modification, e.g., which reduces, e.g., inhibits, DNA
synthesis, 6.3., by a DNA rase.
In an embodiment, the nucleotide modification inhibits a DNA polymerase from extending
the DNA. Without wishing to be bound by , it is ed that in an embodiment any chemical
entity that reduces (e.g., blocks) DNA polymerase extension can be used in accordance with the
methods described herein.
In an embodiment, the nucleotide ation is an insertion of a spacer to the primer, e. g.,
between two adjacent nucleotides in the primer. In an embodiment, the spacer is a flexible spacer. In
an embodiment, the spacer is a carbon spacer (e.g., -(CH2)n-, wherein n=3, 4, 5, 6, 7, 8, 9, 10, or
more), two or more (e.g., three, four, five, six, seven, eight, nine, ten, or more) abasic nucleotides, or a
polyethylene glycol (PEG) spacer. In an ment, the spacer is a PEG spacer. In an embodiment,
the tide ation is 2’-O-methyl, 2’-OH, 2’-NH2, or uracil, e. g., to a ribose.
In an embodiment, the nucleotide modification is located internally or at the 3’ end of the
primer. In an embodiment, at least one primer comprises (i) a first ; (ii) a second member;
and ally (iii) a third member, e.g., comprising a nucleotide modification described herein, e.g.,
located between (i) and (ii).
In an embodiment, the first member is capable of annealing with the second member. In an
embodiment, the first member is capable of annealing with the second member in the same primer,
e.g., through intra-molecular hybridization, e.g., to form a hairpin structure comprising a duplex
region of 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, more basepairs. In another
embodiment, the first member is capable of annealing izing with the second member in a
different primer, e.g., through inter-molecular hybridization, e. g., to form a double-stranded ure
comprising a duplex region of 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, more
basepairs. Without g to be bound by theory, it is believed that in an embodiment, there are at
least two secondary structures that the modified primers can form and facilitate reduction (e.g.,
prevention) of competition to ate (e.g., bead) capture. For example, the secondary structure can
be a hairpin-like structure formed by intra-molecular ization (within the same primer), or the
secondary structure can be a duplex structure formed by molecular hybridization (between two
different primers).
In an embodiment, the first member comprises a sequence that is complementary to the
sequence of an oligonucleotide attached to the capture substrate. In an embodiment, the second
member comprises (e.g., from 5’ to 3’) one, two, or all of: (i) a sequence that is complementary to at
least a n of the first member; (ii) a universal priming sequence (e.g., for PCR amplification or
next-generation sequencing); and (iii) a sequence complementary to a target ce, e. g., an
XCVRS and/or a YCVRS. In an embodiment, the universal priming sequence is identical, or
substantially identical, to the sequence that is complementary to at least a portion of the first member.
In another embodiment, the universal priming sequence is different from the sequence that is
complementary to at least a portion of the first member. In an embodiment, the second member
comprises a sequence for gous ination (e.g., in a yeast or mammalian cell).
In an embodiment, at least one primer comprises a sequence ng at least a portion of a
linker sequence, or a complementary sequence thereof. In an ment, the primer that comprises
a sequence encoding at least a portion of a linker sequence, or a complementary sequence thereof, is
phosphorylated, e.g., 5 ’ phosphorylated. t wishing to be bound by theory, it is believed that in
an embodiment, any sequence with the general properties of flexibility (e.g., facilitated by glycine)
and hydrophilicity can work effectively in accordance with the methods described herein. Exemplary
linkers can generally have overrepresentation of one or more of Gly, Ser, Thr, or Ala and
underrepresentation of hydrophobic residues, e.g., one or more of Trp, Tyr, Phe, Cys, Met, Leu, or Ile.
The length of the primer may vary, e.g., 3-50 amino acid residues (e.g., 5-45, 10-40, 15-35, 20-30, 10-
20, 10-30, 20-40, or 30-40 amino acid residues). In an embodiment, the linker sequence comprises, or
ts of, ((Gly)m-Ser))n, where m=3, 4, 5, or more and n=1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more. In an
embodiment, the linker sequence comprises, or consists of, (Gly-Gly-Gly-Gly-Ser)n, where n=1, 2, 3,
4, 5, 6, 7, 8, 9, 10, or more.
In an ment, the primer is a primer described , e.g., in Examples.
In an embodiment, the reverse transcription, the amplification, or both, occurs in a solution in
the ed production reaction site, e. g., production micro-chamber. In an embodiment, the reverse
transcription, the amplification, or both, does not occur on the substrate (e.g., bead). For example, the
reverse transcription, the amplification, or both, can occur on in a solution within a droplet.
In an embodiment, the XC ds cDNA comprises a 5’ overhang, e.g., a 5’ overhang that is
capable of hybridizing to an oligonucleotide attached to a capture substrate. In an embodiment, the
KC ds cDNA comprises a blunt end, e.g., a blunt end comprising a 5’ phosphate. In an embodiment,
the YC ds cDNA comprises a 5’ overhang, e.g., a 5’ overhang that is capable of hybridizing to an
oligonucleotide attached to a capture substrate. In an embodiment, the YC ds cDNA comprises a
blunt end, 6.3., a blunt end comprising a 5’ phosphate. In an embodiment, the XC ds cDNA and the
YC ds cDNA comprise sticky ends, e.g., both have 5’ overhangs.
In an embodiment, the XC strand and the YC strand are covalently linked, e.g., ligated, to
produce a single stranded nucleic acid sequence, wherein the XC and YC strands are both sense
strands or both antisense strands. In an embodiment, a denatured XC strand of the XC ds cDNA to a
denatured YC strand of the YC ds cDNA are covalently linked, e. g., ligated, n the XC and YC
strands are both sense strands or both antisense strands. In an embodiment, the XC strand is present
in the XC ds cDNA and the YC strand is present in the YC ds cDNA, and wherein the XC ds cDNA
and the YC ds cDNA are covalently linked, e.g., ligated, e.g., to produce a double stranded nucleic
acid ce.
In an embodiment, the covalent linking, e.g., on, occurs in the isolated production
reaction site. In an embodiment, the isolated tion reaction site, e.g., a production micro-
chamber, or the isolated e reaction site, e.g., a linkage chamber, comprises a reagent that
is e of covalently g, e.g., ligating, the XC and YC strands or the XC and YC ds cDNAs.
In an embodiment, the isolated tion reaction site, e.g., a production micro-chamber comprises
an enzyme that covalently couples the XC and YC strands or the XC and YC ds cDNAs. In an
embodiment, the enzyme is a ligase, e.g., a thermal stable . In an embodiment, the covalent
linking comprises ligase thermocycling.
In an embodiment, the covalent linking, e.g., on, occurs in a site different from the
isolated production reaction site, e.g., occurs in an isolated linkage reaction site, e.g., a linkage micro-
chamber. In an embodiment, the XC strand and the YC strand are transferred from the isolated
production site to the isolated linkage reaction site, e.g., a linkage micro-chamber, and the covalent
linking occurs in the isolated linkage reaction site, e.g., a e micro-chamber. In an embodiment,
the isolated linkage reaction site, e.g., a linkage micro-chamber, comprises a reagent that is e of
ntly linking, e.g., ligating, the XC and YC strands or the XC and YC ds cDNAs. In an
embodiment, the isolated linkage reaction site, e.g., a linkage micro-chamber, ses an enzyme
that ntly couples the XC and YC strands or the XC and YC ds cDNAs. In an embodiment, the
enzyme is a ligase, e.g., a thermal stable . In an embodiment, the covalent linking comprises
ligase thermocycling.
In an embodiment, the nt linking, e. g., ligation, comprises: (a) heating the isolated
linkage reaction site, e.g., the linkage micro-chamber, under conditions (e.g., at 950C) that allow
denaturation of the XC strand and the YC strand; (b) cooling the isolated linkage reaction site, e. g.,
the linkage micro-chamber, under conditions (e.g., at 50-65°C) that allow hybridization of the splint
oligonucleotide to the XC strand and the YC strand; (c) maintaining the ed linkage reaction site,
e.g., the linkage micro-chamber, under conditions (e.g., at 45-65°C) that allow ligation of the XC
strand and the YC strand (e.g., formation of phosphodiester bond between the XC strand and the YC
strand); and (d) repeating steps (a), (b), and (c) sequentially for 2, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50,
or more cycles.
In an ment, the XC strand and the YC strand are covalently linked, e.g., ligated, in the
ce of a splint ucleotide. In an embodiment, the splint oligonucleotide is hybridized to a
ce comprising the junction of the XC strand and the YC strand, or a sequence complementary
thereof, and forms a duplexed region at the site of ligation. In an embodiment, the splint
oligonucleotide comprises a modification (e.g., an NHZ group) that inhibits DNA synthesis, e.g., by a
DNA polymerase. In an embodiment, the modification is at the 3’ end of the splint oligonucleotide.
In an ment, a strand complimentary to the covalently linked, e. g., ligated, XC and YC
strands is produced by amplification.
In an embodiment, the , e.g., the step of covalent linkage, does not include a step of
overlap extension polymerase chain reaction (OE-PCR), also known as splicing by overlap extension
or splicing by overhang extension (SOE) PCR.
In an embodiment, the method further ses, prior to acquiring the ed production
reaction site, e.g., a production micro-chamber, acquiring an mRNA loaded capture substrate.
In an embodiment, acquiring the mRNA loaded capture substrate sing: a) acquiring an
isolated cell reaction site, e.g., a cell isolation micro-chamber, comprising: i) a cell; and ii) a capture
substrate capable of binding a first mRNA encoding an XCVR from the cell and a second mRNA
encoding a YCVR from the cell; and b) maintaining the isolated cell on site, e.g., the cell
isolation micro-chamber, under conditions that allow lysis of the cell and binding of the capture
substrate with the first mRNA and the second mRNA to form the mRNA loaded capture substrate,
wherein the isolated cell reaction site, e.g., cell isolation micro-chamber, does not include a nucleic
acid encoding an XCVR or a YCVR from a cell other than the cell (e.g., a different cell).
In an embodiment, the isolated cell reaction site, e. g., cell isolation micro-chamber, comprises
a lysing reagent, e.g., a ent. In an embodiment, the cell is lysed by heat or an enzyme. In an
embodiment, the capture substrate comprises a moiety (e.g., an oligonucleotide) which binds mRNA,
e.g., an oligo(dT).
In an embodiment, the method further ses releasing the mRNA loaded capture
substrate from the isolated cell reaction site, e.g., the cell isolation micro-chamber. In an
embodiment, the releasing step is performed in the presence of a poly(dA) or poly(dT)
oligonucleotide, e.g., to reduce cross-binding of non-captured mRNA.
In an ment, the mRNA loaded capture substrate is transferred from the isolated cell
reaction site, e.g., the cell isolation micro-chamber, to the isolated production reaction site, e.g., the
production micro-chamber.
In an embodiment, the method further comprises releasing the nucleic acid sequence from the
isolated production on site, e.g., the production chamber. In an embodiment, the method
further ses amplifying the nucleic acid sequence. In an embodiment, amplification of the
nucleic acid sequence occurs outside the isolated production reaction site, e.g., the production micro-
r, e. g., after the c acid is released from the isolated production reaction site, e. g., the
production micro-chamber. In an embodiment, amplification of the nucleic acid sequence occurs at
the isolated production reaction site, e.g., the production micro-chamber.
In an embodiment, the method further comprises cing all or a portion of the nucleic
acid sequence.
In an embodiment, the method further comprises inserting all or a portion of nucleic acid
sequence into a vector. In an embodiment, the vector supplies an additional XC element or YC
element not included in the nucleic acid sequence. In an ment, the vector supplies an XC
CDRl, an XC CDR2, or both. In an embodiment, the method further comprises sing the
vector.
In an embodiment, the method further comprises expressing the nucleic acid sequence to
produce a polypeptide sing a segment that encodes an XC element of the XCVR, e.g., an
XCVRS, and a segment that encodes a YC element of the YCVR, e.g., a YCVRS. In an embodiment,
the YC element is N—terminal to the KC element in the ptide. In an embodiment, the XC
t is C-terminal to the YC element in the polypeptide.
In an embodiment, the method r comprises contacting the polypeptide with an antigen.
In an embodiment, the method further comprises determining if the ptide binds the antigen, in
vitro, ex vivo, or in viva, e.g., by a method or assay described herein.
In an aspect, the disclosure features a method of making a nucleic acid sequence comprising a
ce that encodes an X chain element (XC element) of an antibody heavy chain variable region
(XCVR) and a Y chain element (YC element) of an antibody light chain variable region (YCVR), and
wherein the XCVR and YCVR are matched, comprising:
a) acquiring an isolated cell reaction site (e.g., an ed cell on site described herein),
e.g., a cell isolation micro-chamber, comprising: i) a cell (e.g., a cell described herein); and ii) a
capture substrate (e. g., a capture substrate described herein) capable of g a first mRNA
encoding an XCVR from the cell and a second mRNA encoding a YCVR from the cell;
b) maintaining the isolated cell reaction site, e.g., the cell isolation micro-chamber, under
conditions that allow lysis of the cell and binding of the capture substrate with the first mRNA and the
second mRNA to form an mRNA loaded capture substrate,
wherein the isolated cell reaction site, e.g., cell isolation micro-chamber, does not include a
nucleic acid ng an XCVR or a YCVR from a cell other than the cell (e.g., a different cell);
c) contacting the mRNA loaded capture substrate with a on mixture, e.g., a reaction
mixture sing reverse transcriptase, that uses the loaded mRNA as a template to make cDNA
(this can occur, e.g., in the isolated cell reaction site, in an ed production reaction site, or in
neither, e.g., not in an ed reaction site);
(1) acquiring an isolated production reaction site (e.g., an isolated production reaction site
described herein), e.g., a production micro-chamber, comprising: i) an X chain (XC) , n
the XC strand is a strand of an X chain double-stranded cDNA (XC ds cDNA) comprising a segment
that encodes an XC element of the XCVR from the cell, e.g., an X chain variable region sequence
(XCVRS); and ii) a Y chain (YC) strand, wherein the YC strand is a strand of a Y chain double-
stranded cDNA (YC ds cDNA) comprising a segment that encodes a YC element of the YCVR from
the cell, e.g., a Y chain variable region sequence (YCVRS),
wherein the ed production reaction site, e.g., a production micro-chamber, does not
include a nucleic acid encoding a YCVR or an XCVR from a cell other than the cell (e. g., a different
cell); and
e) covalent linking, e. g., ligation, of the XC strand to the YC .
In an embodiment, one or more (e.g., two, three, four, or all) of the steps a)-e) are performed
in accordance with a method described herein. In an embodiment, each of the steps a)-e) is performed
in accordance with a method described .
In an aspect, the disclosure features a method of making a nucleic acid sequence comprising a
sequence that encodes an X chain t (XC element) of an antibody heavy chain variable region
(XCVR) and a Y chain element (YC element) of an antibody light chain variable region (YCVR), and
wherein the XCVR and YCVR are matched, comprising:
a) acquiring an isolated cell reaction site (e.g., an isolated cell reaction site described herein),
e.g., a cell isolation micro-chamber, comprising: i) a cell (e.g., a cell described herein); and ii) a
capture substrate (e.g., a capture substrate described herein) capable of binding a first mRNA
encoding an XCVR from the cell and a second mRNA encoding a YCVR from the cell;
2017/068204
b) maintaining the isolated cell on site, e.g., the cell isolation chamber, under
conditions that allow lysis of the cell and g of the capture substrate with the first mRNA and the
second mRNA to form the mRNA loaded capture substrate,
wherein the isolated cell reaction site, e.g., cell isolation micro-chamber, does not include a
nucleic acid encoding an XCVR or a YCVR from a cell other than the cell (e.g., a different cell);
c) acquiring an ed production reaction site (e.g., an isolated production reaction site
described herein), e. g., a production micro-chamber, comprises: contacting the mRNA loaded capture
substrate with a reaction mixture, e.g., a on e comprising reverse transcriptase, that uses
the loaded mRNA as a template, to produce: a first double-stranded cDNA (ds cDNA) comprising a
strand that is complementary to a first mRNA that encodes an XCVR from a cell; and a second ds
cDNA comprising a strand complementary to a second mRNA encoding a YCVR from the cell (the
cDNA loaded capture substrate);
wherein the isolated tion reaction site, e.g., a production micro-chamber, does not
include a nucleic acid encoding a YCVR or an XCVR from a cell other than the cell (e.g., a different
cell).
(1) maintaining the isolated production reaction site, e.g., the production micro-chamber,
under conditions that allow amplification of the first and second ds cDNAs, to produce: a plurality of
XC ds cDNAs comprising a t that encodes an XC element of the XCVR from the cell, e.g., an
XCVRS; and a plurality of YC ds cDNAs comprising a segment that encodes a YC element of the
YCVR from the cell, e.g., a YCVRS;
e) acquiring an isolated linkage reaction site (e.g., an isolated linkage reaction site described
herein), e. g., a linkage micro-chamber, comprising: covalent linking, e. g., ligation, of a strand of the
KC ds cDNA (XC strand) to a strand of the YC ds cDNA (YC strand), wherein the XC and YC
strands are both sense strands or antisense strands; and
f) amplifying the covalently linked, e.g., ligated, KC and YC strands.
In an embodiment, one or more (e.g., two, three, four, five, or all) of the steps a)-f) are
performed in accordance with a method described herein. In an embodiment, each of the steps a)-f) is
performed in accordance with a method described herein.
In an aspect, the disclosure features a method of making a library comprising a plurality of
unique s, the method comprising:
making the plurality of members, n each of the members comprises a ce that
encodes an X chain element (XC element) of an X chain variable region (XCVR) and a Y chain
element (YC element) of a Y chain variable region (YCVR), and wherein the XCVR and YCVR are
matched, made by a method described ,
wherein each unique nucleic acid sequence of the plurality comprises an XC element and a
YC element from a different unique cell (e.g., a cell bed herein),
thereby making a library sing a plurality of unique members.
In an embodiment, the plurality of unique members comprises at least 104, 105, 106, 107, 108,
or 109 unique members. In an embodiment, the plurality of unique members comprises 104 to 109, 104
to 108, 104 to 107, 104 to 106, 104 to 105, 108 to 109, 107 to 109, 106 to 109, 105 to 109, 105 to 108, 106 to
107, 104 to 105, 105 to 106, 106 to 107, 107 to 108, or 108 to 109 unique members. In an embodiment, at
least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%, of the members in the library are unique
members (which encode matched XC element and YC elements sequences). In an embodiment, less
than 20%, 15%, 10%, 5%, 4%, 3%, 2%, or 1%, of the s in the library are unique members
(which encode matched XC element and YC elements sequences).
In an aspect, the disclosure es a library comprising a ity of unique members,
wherein,
i) each unique member of the plurality comprises a segment that encodes an XC element, e.g.,
an XCVRS, and a segment that encodes a YC element, e.g., a YCVRS, wherein the XC element and
the YC element in each unique member is matched;
ii) each unique member of the plurality comprises a segment that encodes an XC element,
e.g., an XCVRS, and a t that encodes a YC element, e.g., a YCVRS, from a different unique
cell; and
iii) the library comprises one or more (e.g., two, three, four, or all) of the following
properties:
a) the library is made by a method described herein;
b) the plurality of unique members comprises at least 104, 105, 106, 107, 108, or 109
unique nucleic acid sequences;
c) the plurality of unique s comprises 104 to 109, 104 to 108, 104 to 107, 104 to
106, 104 to 105, 108 to 109, 107 to 109, 106 to 109, 105 to 109, 105 to 108, 106 to 107, 104 to 105,
105 to 106, 106 to 107, 107 to 108, or 108 to 109 unique s;
d) at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%, of the members in
the library are unique members (which encode matched XC element and YC elements
sequences); or
e) less than 20%, 15%, 10%, 5%, 4%, 3%, 2%, or 1%, of the members in the library
are unique members (which encode matched XC element and YC elements sequences).
In an embodiment, each unique member of the plurality is red such that, when
expressed, the XC element, e. g., the XCVRS, and the YC element, e.g., the YCVRS, form a
functional antigen binding molecule, e.g., an scFV.
In an embodiment, the library is a display library. In an embodiment, each of the members of
the ity further encodes a polypeptide that results in display of the member on the surface of a
2017/068204
display . In an ment, the library is a phage display library. In an embodiment, the
library is a yeast display library. In an embodiment, the library is a mammalian display library.
In an aspect, the disclosure features a method of making a binding polypeptide (e.g., a
polypeptide comprising an XC element and a YC element), the method comprising: a) acquiring a
library described herein, e.g., by a method described ; and b) expressing a polypeptide encoded
by a unique nucleic acid of the library.
In an embodiment, the method further comprises contacting the polypeptide with an antigen.
In an embodiment, the method r comprises retrieving (e.g., isolating or purifying) the nucleic
acid that encodes a polypeptide that binds the antigen.
In an aspect, the disclosure es an isolated production reaction site, e.g., a production
micro-chamber, which is an isolated production reaction site described herein (e.g., comprising a
nucleic acid ng an XCVR and a nucleic acid ng a YCVR, wherein the XCVR and the
YCVR are matched).
In an ment, the isolated production reaction site, e.g., a production micro-chamber,
does not include a nucleic acid encoding an XCVR or a YCVR from a different cell.
In an embodiment, the isolated production reaction site, e.g., a tion micro-chamber,
comprises one, two, or all of: (i) one or more primers specific to V gene sequences of the XC and YC;
(ii) one or more primers specific to overhangs introduced onto the XC and YC cDNAs; or (iii) one or
more primers comprising a first member, a second member, and a third member comprising a
nucleotide ation (e.g., a spacer) located between the first and second members, wherein the
first member is capable of annealing with the second member of the same primer or a different
primer, e.g., forming a structure sing a duplex region of 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15,
l6, l7, l8, 19, 20, more basepairs.
In an embodiment, the isolated production reaction site, e.g., a production micro-chamber,
does not comprise a reagent that can covalently link nucleic acids, e.g., a ligase, e.g., a thermostable
ligase. In another embodiment, the isolated production reaction site, e. g., a production micro-
chamber, comprises a reagent that can covalently link nucleic acids, e.g., a ligase, e.g., a thermostable
ligase.
In an aspect, the sure features a self-annealing oligonucleotide comprising a first
member, a second member, and third member sing a nucleotide modification (e. g., a spacer)
located between the first and second members, wherein the first member is capable of annealing with
the second member of the same oligonucleotide (e.g., for a method of making a c acid sequence
comprising a sequence that encodes an XC element of an XCVR and a YC element of a YCVR,
wherein the XCVR and YCVR are matched).
In an ment, the first and second members are capable of forming a n structure
comprising a duplex region of 4, 5,6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, more
basepairs. In an embodiment, the first member is 5-40 tides, e. 3., 5-10, 5-20, 5-30, 30-40, 20-
40, 10-30, 10-30, or 15-25 nucleotides, in length. In an embodiment, the second member is 5-40
nucleotides, e.g., 5-10, 5-20, 5-30, 30-40, 20-40, 10-30, 10-30, or 15-25 nucleotides, in length.
In an embodiment, the spacer is a spacer described herein, e.g., a flexible spacer or a PEG
spacer.
In an embodiment, the first member ses a sequence that is complementary to the
sequence of an oligonucleotide attached to a capture substrate.
In an embodiment, the second member comprises (e.g., from 5’ to 3’) one, two, or all of: (i) a
sequence that is complementary to at least a portion of the first member; (ii) a universal priming
sequence (e.g., for PCR amplification or next-generation sequencing); and (iii) a sequence
complementary to a target sequence, e.g., an XCVRS and/or a YCVRS. In an embodiment, the
universal priming sequence is identical, or substantially identical, to the ce that is
complementary to at least a portion of the first member. In another embodiment, the universal
priming sequence is different from the sequence that is complementary to at least a portion of the first
. In an embodiment, the second member comprises a ce for homologous
recombination (e.g., in a yeast or mammalian cell).
In an aspect, the disclosure features an isolated linkage reaction site, e. g., a e micro-
r, which is an isolated linkage reaction site bed herein (e.g., comprising a c acid
encoding an XCVR and a c acid encoding a YCVR, wherein the XCVR and the YCVR are
matched).
In an ment, the isolated linkage reaction site, e.g., a linkage micro-chamber, does not
include a nucleic acid encoding an XCVR or a YCVR from a different cell.
In an embodiment, the isolated linkage reaction site, e.g., a linkage micro-chamber, comprises
a splint oligonucleotide (e.g., a splint oligonucleotide described herein) that is capable of hybridizing
to a sequence comprising the junction of the XC strand and the YC strand, or a sequence
complementary thereof, to form a duplexed region at the site of ligation.
In an embodiment, the isolated e reaction site, e.g., a e chamber, comprises
a reagent that can covalently link nucleic acids, e.g., a ligase, e.g., a thermostable ligase.
T-Cell Receptor Molecules
The disclosures herein are not intended to be limited to antibody molecules. In an
embodiment, the binding molecule is a TCR molecule, e. g., a soluble TCR molecule. In an
embodiment, the binding molecule comprises a TCR or chain variable region and a TCR [3 chain
variable region. In an embodiment, the binding molecule comprises a TCR 7 chain variable region
and a TCR 5 chain variable region. For example, in any of the aspects, embodiments, and definitions
herein, an dy heavy chain (or variable region) can be replaced with a TCR or chain (or variable
region), and an antibody light chain (or variable region) can be replaced with a TCR [3 chain (or
variable ); or an antibody heavy chain (or variable region) can be replaced with a TCR 7 chain
(or variable region), and an antibody light chain (or variable ) can be replaced with a TCR 5
chain (or variable region).
Disclosed herein are T-cell receptor (TCR) molecules and libraries of TCR molecules. In an
embodiment, the TCR molecule or library of TCR molecules are made by a method described herein.
As used herein, the term “TCR molecule,” also known as “T-cell receptor molecule” or “T
cell or molecule,” refers to a protein, e.g., a TCR chain or a nt thereof, comprising at
least one TCR variable domain sequence. The term “TCR molecule” includes, for example, full-
length, mature TCRs and antigen-binding nts of a TCR. For e, a TCR molecule can
include an 0t chain variable domain sequence and a B chain variable domain ce. In another
example, a TCR molecule can e a y chain variable domain sequence and a 6 chain variable
domain sequence. In an embodiment, the TCR molecule is a soluble TCR molecule.
T-cell receptors can be found on the surface of T cells and are responsible for izing
fragments of antigen as peptides bound to major histocompatibility complex (MHC) molecules. A
TCR typically include two different protein chains. In humans, in about 95% of T cells the TCR
include an alpha (or) chain and a beta ([3) chain (encoded by TRA and TRB, respectively), whereas in
about 5% of T cells the TCR include gamma and delta (7/5) chains (encoded by TRG and TRD,
respectively). This ratio can change during ontogeny and in diseased states (such as leukemia).
When the TCR engages with antigenic peptide and MHC (peptide/MHC), the T lymphocyte is
ted through signal transduction, e.g., a series of biochemical events mediated by associated
enzymes, co-receptors, specialized adaptor molecules, and activated or released transcription factors.
For example, the lly-occurring TCR is typically a disulfide-linked membrane-anchored
heterodimeric protein including, e.g., the highly variable alpha (or) and beta ([3) chains expressed as
part of a complex with the invariant CD3 chain molecules. T cells expressing this receptor are
sometimes referred to as azfi (or (XB) T cells, though a ty of T cells s an alternate receptor,
formed by variable gamma (y) and delta (6) chains, sometimes referred as 1/5 T cells.
Each chain of TCR can include two extracellular domains: a variable (V) region and a
constant (C) region. The constant region is proximal to the cell membrane, followed by a
transmembrane region and a short cytoplasmic tail, while the variable region can bind to the
peptide/MHC complex.
The variable domain of the TCR or chain or [5 chain each can have three hypervariable or
complementarity ining regions (CDRs). There can also be an additional area of
hypervariability on the [5 chain (HV4), which typically does not contact antigen and is not considered
a CDR. The es in these variable domains are located in two regions of the TCR, at the interface
of the or and [3 chains and in the [3-chain framework region that is in proximity to the CD3 -
transduction complex. Without wishing to be bound by theory, it is believed that in an embodiment,
CDR3 is the main CDR responsible for recognizing processed antigen. CDRl of the alpha chain can
interact with the N—terminal part of the antigenic peptide, and CDRl of the [3 chain can interact with
the C-terminal part of the peptide. CDR2 may ize the MHC. CDR4 of the [3 chain is generally
not thought to participate in antigen recognition, but may interact with superantigens.
The constant domain of the TCR include, e.g., short connecting sequences, which form a link
between the two chains, e.g., through de bonds.
The generation of TCR diversity arises mainly from c recombination of the DNA
encoded segments in individual somatic T cells by somatic V(D)J recombination . Each recombined
TCR may possess unique antigen specificity, determined by the structure of the antigen-binding site,
e.g., formed by the 0t and [3 chains in case of (113 T cells or y and 5 chains on case of 1/5 T cells. For
example, the TCR 0c and y chains can be generated by VJ recombination, and the [3 and 5 chains can
be generated by VDJ recombination. The intersection of these specific regions (e.g., V and J for 0t or
7 chain; V, D, and J for [3 or 5 chain) corresponds to the CDR3 region that is typically important for
peptide/MHC recognition.
The TCR receptor can form a complex of variable TCR chains (e.g., 0t and [3 chains with three
dimeric signaling modules CD35/e, CD3~yle and CD247 C/C or Cjn). T cell can express clonal TCRs
which ize specific peptide/MHC complex during al contact n T cell and antigen-
ting cell-APC (MHC class II) or any other cell type (MHC class I). The signal from the T-cell
complex can be enhanced by simultaneous binding of the MHC molecules by a specific co-receptor.
For example, on helper T cells and regulatory T cells, the co-receptor is CD4 that is ic for MHC
class II, and on cytotoxic T cells, the co-receptor is CD8 that is specific for MHC class I.
The term “TCR” includes intact molecules as well as functional fragments thereof. TCR
fragments may be obtained using any suitable method, including several conventional techniques
known to those with skill in the art, and the fragments can be screened for utility in the same manner
as are intact TCRs. Constant regions of the TCRs can be altered, e.g., mutated, to modify the
properties of the TCR.
The TCR molecule can be a single chain TCR. The single chain TCR can be dimerized or
multimerized to generate alent TCRs having specificities for different epitopes of the same
target protein.
The TCR les disclosed herein can also be single domain TCRs. Single domain TCRs
can include TCRs whose complementary determining regions are part of a single domain ptide.
es e, but are not limited to, or, [3, y, or 5 chain TCRs, TCRs naturally devoid of a 0t, [3, y,
or 5 chain, single domain TCRs derived from conventional two-chain TCRs, engineered TCRs and
single domain scaffolds other than those derived from TCRs. Single domain TCRs may be any of the
2017/068204
art, or any future single domain TCRs. Single domain TCRs may be derived from any species
including, but not limited to mouse, human, camel, llama, fish, shark, goat, rabbit, and bovine.
The variable regions can be subdivided into regions of hypervariability, termed
“complementarity determining regions” (CDR), interspersed with regions that are more conserved,
termed “framework s” (FR or FW). The terms “complementarity determining region,” and
“CDR,” as used herein in the context of TCR molecules refer to the ces of amino acids within
TCR variable regions which confer antigen specificity and binding affinity. As used herein, the terms
“framework,” “FW” and “FR” are used interchangeably.
As used herein, a “TCR variable domain sequence” refers to an amino acid sequence which
can form the structure of a TCR variable domain. For example, the sequence may include all or part
of the amino acid sequence of a naturally-occurring variable domain. For example, the sequence may
or may not include one, two, or more N— or C-terminal amino acids, or may include other alterations
that are compatible with formation of the protein structure.
The term “antigen-binding region” refers to the part of a TCR molecule that comprises
determinants that form an interface that binds to an antigen, or an epitope thereof. With respect to
proteins (or protein mimetics), the antigen-binding region lly includes one or more loops (of at
least, e.g., four amino acids or amino acid mimics) that form an ace that binds to the antigen.
Typically, the antigen-binding region of a TCR molecule includes at least one or two CDRs and/or
hypervariable loops, or more lly at least three, four, five or six CDRs and/or hypervariable
loops.
The terms “compete” or “cross-compete” are used interchangeably herein to refer to the
ability of a TCR molecule to ere with binding of r TCR molecule, to a target. The
interference with g can be direct or indirect (e.g., through an allosteric modulation of the TCR
molecule or the target). The extent to which an antibody molecule is able to interfere with the binding
of another TCR molecule to the target, and therefore whether it can be said to compete, can be
determined using a competition binding assay, for example, a FACS assay, an ELISA or BIACORE
assay. In an embodiment, a competition binding assay is a quantitative competition assay. In an
embodiment, a first TCR molecule is said to compete for g to the target with a second TCR
molecule when the g of the first antibody molecule to the target is reduced by 10% or more,
e.g., 20% or more, 30% or more, 40% or more, 50% or more, 55% or more, 60% or more, 65% or
more, 70% or more, 75% or more, 80% or more, 85% or more, 90% or more, 95% or more, 98% or
more, 99% or more in a competition binding assay (e.g., a competition assay described herein).
The TCR molecule can be a onal or a monoclonal. In some embodiments, the TCR can
be recombinantly ed, e.g., produced by any suitable phage display or combinatorial methods.
Phage display and combinatorial methods are known in the art.
In an embodiment, the TCR molecule is a fully human TCR (e. g., a TCR made in a mouse
which has been genetically engineered to produce a TCR from a human TCR sequence), or a non-
human TCR, e.g., a rodent (mouse or rat), goat, primate (e.g., monkey), camel TCR. In an
embodiment, the man TCR is a rodent (mouse or rat TCR). For example, Human TCRs can be
generated using transgenic mice carrying the human TCR genes rather than the mouse system.
A TCR can be one in which the variable region, or a portion thereof, e.g., the CDRs, are
generated in a non-human organism, e.g., a rat or mouse. Chimeric, CDR-grafted, and zed
TCRs are within the invention. TCRs generated in a non-human organism, e.g., a rat or mouse, and
then d, e.g., in the variable framework or constant region, to decrease antigenicity in a human
are within the invention.
Chimeric TCRs can be produced by any suitable recombinant DNA technique.
A humanized or CDR-grafted TCR will have at least one or two but generally all three
recipient CDRs (of TCR chains) replaced with a donor CDR. The TCR may be replaced with at least
a portion of a non-human CDR or only some of the CDRs may be replaced with non-human CDRs. It
is only necessary to replace the number of CDRs required for binding of the zed TCR to an
antigen. In an embodiment, the donor will be a rodent TCR, e.g., a rat or mouse TCR, and the
recipient will be a human framework or a human consensus framework. Typically, the TCR
providing the CDRs is called the “donor” and the TCR providing the framework is called the
“acceptor.” In some embodiments, the donor TCR is a non-human (e.g., rodent). The acceptor
framework is typically a naturally-occurring (e.g., a human) framework or a consensus framework, or
a ce about 85% or higher, e.g., 90%, 95%, 99% or higher identical thereto.
As used herein, the term “consensus sequence” refers to the sequence formed from the most
ntly occurring amino acids (or nucleotides) in a family of related sequences (See e.g., er,
From Genes to Clones (Verlagsgesellschaft, Weinheim, Germany 1987). In a family of proteins, each
position in the consensus sequence is occupied by the amino acid occurring most frequently at that
position in the family. If two amino acids occur equally frequently, either can be ed in the
consensus ce. A “consensus framework” refers to the framework region in the consensus TCR
sequence.
A TCR can be humanized by any suitable method. Humanized or CDR-grafted TCR s can be
produced by afting or CDR substitution, wherein one, two, or all CDRs of an immunoglobulin
chain can be replaced. Also provided are humanized TCRs in which specific amino acids have been
substituted, deleted or added.
In an embodiment, the TCR molecule has a constant region. The nt region can be
altered, e.g., mutated, to modify a property of the TCR molecule. In an embodiment, a constant
region of the TCR molecule is altered. Methods for altering a constant region are known in the art.
In an embodiment, the only amino acids in the TCR le are cal amino acids. In
an embodiment, the TCR molecule comprises naturally-occurring amino acids; s, derivatives
and congeners thereof; amino acid analogs having variant side chains; and/or all stereoisomers of any
of any of the foregoing. The TCR molecule may comprise the D- or L— optical isomers of amino
acids and peptidomimetics.
A polypeptide of a TCR molecule described herein may be linear or branched, it may
comprise modified amino acids, and it may be interrupted by non-amino acids. The TCR molecule
may also be modified; for example, by disulfide bond formation, glycosylation, lipidation, ation,
phosphorylation, or any other manipulation, such as conjugation with a labeling component. The
polypeptide can be isolated from natural sources, can be a produced by recombinant techniques from
a eukaryotic or prokaryotic host, or can be a product of synthetic procedures.
The TCR molecule described herein can be used alone in unconjugated form, or can be bound
to a substance, e.g., a toxin or moiety (e.g., a therapeutic drug; a nd emitting radiation;
molecules of plant, fungal, or bacterial origin; or a biological protein (e. g., a protein toxin) or le
(e. g., a recombinant viral particle, e.g., via a viral coat protein). For example, the TCR molecule can
be coupled to a radioactive isotope such as an (1-, [3-, or y—emitter, or a B-and ter.
A TCR le can be derivatized or linked to another functional molecule (e.g., another
peptide or n). As used herein, a “derivatized” TCR molecule is one that has been modified.
Methods of derivatization include but are not limited to the addition of a fluorescent moiety, a
radionucleotide, a toxin, an enzyme or an affinity ligand such as . ingly, the TCR
molecules are ed to include derivatized and ise modified forms of the dies
described herein, including immunoadhesion molecules. For example, a TCR molecule can be
functionally linked (by chemical coupling, genetic , noncovalent association or otherwise) to
one or more other molecular entities, such as another TCR (e.g., a bispecific TCR), a detectable agent,
a toxin, a pharmaceutical agent, and/or a protein or peptide that can mediate association of the TCR or
TCR portion with another molecule (such as a streptavidin core region or a polyhistidine tag).
Some types of derivatized TCR molecule are produced by crosslinking two or more TCRs (of
the same type or of different types, e.g., to create bispecific TCRs). Suitable inkers include
those that are heterobifunctional, having two distinctly reactive groups separated by an appropriate
spacer (e.g., m—maleimidobenzoyl-N-hydroxysuccinimide ester) or homobifunctional (e.g.,
disuccinimidyl suberate). Such linkers are ble from Pierce Chemical Company, Rockford, Ill.
Useful detectable agents with which a TCR molecule may be tized (or labeled) to
include fluorescent compounds, various enzymes, prosthetic , luminescent materials,
bioluminescent materials, fluorescent emitting metal atoms, e.g., um (Eu), and other anthanides,
and radioactive materials (described below). Exemplary fluorescent detectable agents include
fluorescein, fluorescein isothiocyanate, rhodamine, 5dimethylamine-l-napthalenesulfonyl chloride,
phycoerythrin and the like. A TCR may also be derivatized with detectable enzymes, such as alkaline
phosphatase, adish peroxidase, B-galactosidase, acetylcholinesterase, glucose oxidase and the
like. When a TCR is derivatized with a detectable enzyme, it is ed by adding additional reagents
that the enzyme uses to produce a detectable reaction product. For example, when the detectable
agent horseradish peroxidase is present, the addition of hydrogen peroxide and diaminobenzidine
leads to a colored reaction product, which is detectable. A TCR molecule may also be tized
with a prosthetic group (e.g., streptavidin/biotin and avidin/biotin). For example, a TCR may be
derivatized with biotin, and detected h indirect measurement of avidin or streptavidin binding.
Examples of suitable fluorescent als include umbelliferone, fluorescein, fluorescein
isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl de or phycoerythrin; an
example of a scent material includes luminol; and examples of bioluminescent als
include luciferase, luciferin, and aequorin.
Labeled TCR molecules can be used, for example, diagnostically and/or experimentally in a
number of contexts, including (i) to isolate a predetermined antigen by standard techniques, such as
affinity chromatography or immunoprecipitation; (ii) to detect a predetermined antigen (e.g., in a
cellular lysate or cell supernatant) in order to evaluate the abundance and n of expression of the
n; (iii) to monitor protein levels in tissue as part of a clinical testing procedure, e.g., to determine
the efficacy of a given treatment regimen.
A TCR molecule may be conjugated to another molecular , typically a label or a
therapeutic (e.g., antimicrobial (e.g., antibacterial or icidal), immunomodulatory,
stimularoty, cytotoxic, or cytostatic) agent or moiety. Radioactive isotopes can be used in
diagnostic or therapeutic applications. Radioactive isotopes that can be coupled to the antibody
molecules include, but are not limited to (1-, [3-, or y—emitters, or B-and y—emitters. Such radioactive
isotopes include, but are not limited to iodine (1311 or I), yttrium (90Y), lutetium (177Lu), actinium
(225Ac), praseodymium, astatine (leAt), rhenium (186Re), bismuth (212Bi or 213Bi), indium (1 1 1In),
technetium (99mTc), phosphorus (32F), rhodium (lgth), sulfur (35$) carbon (14C), tritium (3H),
chromium (SlCr), chlorine (36Cl), cobalt (“Co or 58C0), iron (59Fe), selenium (7586), or m (67Ga).
Radioisotopes useful as therapeutic agents include yttrium (90Y), lutetium (177Lu), actinium (225Ac),
praseodymium, astatine , rhenium (186Re), bismuth (212Bi or 213Bi), and rhodium (lgth).
Radioisotopes useful as , e.g., for use in diagnostics, include iodine (1311 or 125I), indium (mln),
technetium (99mTc), phosphorus (32F), carbon (14C), and tritium (3H), or one or more of the therapeutic
isotopes listed above.
The present disclosure provides radiolabeled TCR molecules and methods of labeling the
same. In an embodiment, a method of labeling a TCR molecule is sed. The method includes
contacting a TCR le, with a chelating agent, to y produce a conjugated TCR. The
conjugated antibody is radiolabeled with a radioisotope, e.g., 111Indium, ium and 177Lutetium, to
thereby e a labeled TCR molecule.
In some aspects, this disclosure es a method of making a TCR molecule disclosed
. The method includes: providing an antigen, or a fragment thereof; obtaining a TCR molecule
that specifically binds to the antigen; evaluating efficacy of the TCR molecule in modulating activity
of the antigen and/or organism expressing the antigen. The method can further include administering
the TCR molecule, including a derivative f (e.g., a humanized TCR molecule) to a subject, e.g.,
a human.
This disclosure provides an isolated nucleic acid molecule encoding the above TCR molecule,
vectors and host cells thereof. The c acid le includes, but is not limited to, RNA,
genomic DNA and cDNA.
Animal Models
The polypeptides (e.g., binding polypeptides, e.g., antibody les or TCR molecules)
bed herein can be evaluated in viva, e.g., using various animal models. For example, an animal
model can be used to test the efficacy of a binding polypeptide (e.g., an antibody molecule or TCR
molecule) described herein in modulating a biological function of a target molecule or cell. As
another example, an animal model can also be used to test the efficacy of a binding polypeptide (e.g.,
an antibody molecule or TCR molecule) described herein in in treating, preventing, or diagnosing a
disorder described herein. Animal models can also be used, e.g., to investigate for side effects,
e concentrations of binding polypeptides (e.g., antibody molecules or TCR molecules) in situ,
demonstrate correlations between a function of a target molecule or cell and a disorder described
herein.
Exemplary animal models for other disorders described herein are also known in the art.
ary types of animals that can be used to evaluate the binding ptides (e.g., antibody
molecules or TCR molecules) described herein include, but are not limited to, mice, rats, rabbits,
guinea pigs, and monkeys.
Pharmaceutical Compositions and Kits
In some aspects, this sure provides compositions, e.g., ceutically acceptable
compositions, which include a polypeptide (e.g., a binding polypeptide, e.g., an antibody molecule or
a TCR molecule) bed herein, formulated together with a pharmaceutically acceptable carrier.
As used herein, “pharmaceutically able carrier” includes any and all solvents,
dispersion media, isotonic and absorption delaying agents, and the like that are physiologically
compatible. The r can be suitable for intravenous, intramuscular, subcutaneous, parenteral,
rectal, spinal or epidermal administration (e.g., by injection or on). In certain embodiments, less
than about 5%, e.g., less than about 4%, 3%, 2%, or 1% of the binding polypeptides in the
pharmaceutical composition are present as aggregates. In other embodiments, at least about 95%,
e.g., at least about 96%, 97%, 98%, 98.5%, 99%, 99.5%, 99.8%, or more of the binding polypeptides
in the ceutical composition are present as monomers. In some embodiments, the level of
aggregates or monomers is determined by chromatography, e.g., high performance size exclusion
chromatography (HP-SEC).
WO 19402 2017/068204
The compositions set out herein may be in a variety of forms. These include, for example,
liquid, semi-solid and solid dosage forms, such as liquid solutions (e.g., injectable and infusible
ons), dispersions or suspensions, liposomes, and suppositories. A suitable form depends on the
intended mode of administration and therapeutic application. Typical suitable compositions are in the
form of injectable or infusible solutions. One suitable mode of administration is parenteral (e.g.,
intravenous, aneous, intraperitoneal, intramuscular). In an embodiment, the binding
polypeptide is administered by intravenous infusion or injection. In another embodiment, the binding
polypeptide is administered by intramuscular or aneous injection.
The phrases “parenteral administration” and “administered parenterally” as used herein means
modes of administration other than enteral and topical administration, usually by injection, and
includes, without tion, intravenous, intramuscular, intraarterial, intrathecal, apsular,
intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, aneous, subcuticular,
intraarticular, sular, subarachnoid, intraspinal, epidural and intrasternal injection and infusion.
Therapeutic compositions typically should be sterile and stable under the conditions of
manufacture and storage. The composition can be formulated as a solution, microemulsion,
dispersion, liposome, or other ordered structure suitable to high concentrations of binding
polypeptides (e.g., antibody molecules or TCR molecules). Sterile injectable ons can be
prepared by incorporating the active compound (e.g., binding polypeptide) in the required amount in
an appropriate solvent with one or a combination of ingredients enumerated above, as ed,
followed by filtered sterilization. Generally, dispersions are prepared by incorporating the active
compound into a sterile vehicle that contains a basic dispersion medium and the required other
ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile
injectable ons, the preferred methods of preparation are vacuum drying and freeze-drying that
yields a powder of the active ient plus any onal desired ingredient from a previously
sterile-filtered solution thereof. The proper fluidity of a solution can be maintained, for example, by
the use of a coating such as in, by the maintenance of the required particle size in the case of
dispersion and by the use of surfactants. Prolonged absorption of injectable compositions can be
t about by including in the composition an agent that delays absorption, for example,
monostearate salts and gelatin.
The binding polypeptides (e.g., antibody molecules or TCR receptors) bed herein can be
administered by a variety of s. Several are known in the art, and for many therapeutic,
prophylactic, or stic applications, an appropriate route/mode of administration is intravenous
injection or infusion. For example, the g polypeptides can be administered by intravenous
infusion at a rate of less than lOmg/min; preferably less than or equal to 5 mg/min to reach a dose of
about 1 to 100 mg/mz, preferably about 5 to 50 mg/m2, about 7 to 25 mg/m2 and more ably,
about 10 mg/m2. As will be appreciated by the skilled artisan, the route and/or mode of
administration will vary depending upon the desired results. In certain embodiments, the active
compound may be prepared with a r that will protect the compound against rapid release, such
as a controlled release formulation, ing implants, transdermal patches, and microencapsulated
delivery s. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate,
polyanhydrides, polyglycolic acid, collagen, thoesters, and polylactic acid. Many methods for
the preparation of such formulations are patented or generally known to those skilled in the art. See,
e.g., Sustained and Controlled Release Drug Delivery Systems, J. R. Robinson, ed., Marcel Dekker,
Inc., New York, 1978.
In certain embodiments, the binding polypeptide (e. g., antibody molecule or TCR molecule)
can be orally administered, for e, with an inert diluent or an assimilable edible r. The
binding polypeptide (and other ingredients, if desired) may also be enclosed in a hard or soft shell
gelatin capsule, compressed into tablets, or incorporated directly into the subject’s diet. For oral
therapeutic administration, the binding polypeptide may be incorporated with excipients and used in
the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers,
and the like. To administer the binding polypeptide (e. g., antibody le) by other than parenteral
stration, it may be necessary to coat the compound with, or inister the compound with,
a material to prevent its inactivation. eutic, lactic, or diagnostic compositions can also
be administered with medical devices, and several are known in the art.
Dosage regimens are adjusted to provide the desired response (e. g., a therapeutic,
prophylactic, or diagnostic response). For example, a single bolus may be administered, several
d doses may be administered over time or the dose may be proportionally reduced or increased
as indicated by the exigencies of the eutic situation. It is especially advantageous to formulate
parenteral compositions in dosage unit form for ease of administration and uniformity of dosage.
Dosage unit form as used herein refers to physically discrete units suited as y dosages for the
subjects to be treated; each unit contains a predetermined quantity of active compound calculated to
e the desired therapeutic effect in association with the required pharmaceutical carrier. The
specification for the dosage unit forms are dictated by and directly dependent on (a) the unique
characteristics of the antibody molecule and the particular therapeutic, prophylactic, or diagnostic
effect to be achieved, and (b) the limitations inherent in the art of compounding such a g
polypeptide for the ent of sensitivity in individuals.
An exemplary, non-limiting range for a therapeutically, prophylactically, or diagnostically
effective amount of a binding polypeptide (e. g., an antibody molecule or TCR molecule) is about 0.1-
50 mg/kg, e.g., about 0.1-30 mg/kg, e.g., about 1-30, 1-15, 1-10, 1-5, 5-10, or 1-3 mg/kg, e.g., about
1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 30, 40, or 50 mgikg. The binding polypeptide can be administered
by intravenous infusion at a rate of less than 10 mg/min, e. g., less than or equal to 5 mg/min to reach a
dose of about 1 to 100 mg/mz, e.g., about 5 to 50 mg/m2, about 7 to 25 mg/m2, e.g., about 10 mg/m2.
It is to be noted that dosage values may vary with the type and severity of the ion to be
alleviated. It is to be further understood that for any particular subject, specific dosage regimens
should be adjusted over time according to the individual need and the professional judgment of the
person administering or ising the stration of the compositions, and that dosage ranges set
forth herein are exemplary only and are not intended to limit the scope or practice of the claimed
compositions.
The pharmaceutical compositions herein may e a “therapeutically effective amount,”
“prophylactically ive amount,” or ostically effectively amount” of a binding polypeptide
(e. g., an antibody molecule or TCR molecule) described herein.
A peutically effective amount” refers to an amount effective, at dosages and for periods
of time necessary, to achieve the desired therapeutic result. A therapeutically effective amount of the
binding polypeptide (e.g., antibody molecule or TCR le) may vary according to factors such as
the disease state, age, sex, and weight of the individual, and the ability of the antibody or antibody
portion to elicit a desired response in the individual. A therapeutically effective amount is also one in
which any toxic or detrimental effect of the antibody molecule is outweighed by the therapeutically
ial effects.
A peutically effective dosage” typically inhibits a measurable parameter by at least
about 20%, e.g., by at least about 40%, by at least about 60%, or by at least about 80% relative to
untreated subjects. The measurable parameter may be, e.g., hematuria, colored urine, foamy urine,
pain, swelling (edema) in the hands and feet, or high blood pressure. The ability of a binding
ptide (e.g., an antibody molecule) to t a measurable parameter can be evaluated in an
animal model system predictive of efficacy in treating or preventing a disorder described .
Alternatively, this property of a composition can be evaluated by examining the ability of the binding
polypeptide (e.g., antibody molecule or TCR molecule) to modulate a biological function of a target
molecule or cell, e.g., by an in vitro assay.
A “prophylactically effective amount” refers to an amount effective, at dosages and for
periods of time necessary, to achieve the desired prophylactic result. lly, since a prophylactic
dose is used in subjects prior to or at an earlier stage of e, the prophylactically effective amount
will be less than the therapeutically effective amount.
A “diagnostically effective amount” refers to an amount effective, at dosages and for periods
of time necessary, to achieve the desired diagnostic result. lly, a diagnostically effective
amount is one in which a er, e.g., a disorder described herein, can be sed in vitro, ex vivo,
or in viva.
Also within this sure is a kit that comprises a binding polypeptide (e. g., an antibody
molecule or TCR molecule), described herein. The kit can include one or more other elements
including: instructions for use; other reagents, e.g., a label, a therapeutic agent, or an agent useful for
chelating, or otherwise coupling, an antibody molecule to a label or therapeutic agent, or a
radioprotective composition; devices or other materials for preparing the binding polypeptide (e.g.,
antibody molecule or TCR molecule) for administration; pharmaceutically acceptable carriers; and
devices or other materials for administration to a subject.
Nucleic Acids
The present disclosure also features nucleic acids comprising nucleotide sequences that
encode polypeptides (e. g., binding polypeptides, e.g., antibody molecules or T cell receptor
molecules), as described herein.
In an embodiment, the nucleic acid further ses a nucleotide sequence encoding a heavy
chain variable region of a polypeptide (6.3., an antibody molecule or TCR molecule) bed herein,
or having a nucleotide sequence substantially homologous thereto (e.g., a sequence at least about
85%, 90%, 95%, 99% or more identical thereto, and/or capable of hybridizing under the stringency
conditions described herein). In another embodiment, the nucleic acid further comprises a nucleotide
sequence encoding a light chain le region of a polypeptide (e.g., an antibody molecule or TCR
molecule) described herein, or a nucleotide sequence substantially homologous o (e.g., a
sequence at least about 85%, 90%, 95%, 99% or more identical thereto, and/or capable of hybridizing
under the stringency conditions described herein). In yet another embodiment, the nucleic acid
further comprises a nucleotide sequence encoding a heavy chain le region and a light chain
variable region of a polypeptide (e.g., an antibody molecule or TCR molecule) described herein, or a
nucleotide sequence substantially homologous o (e.g., a sequence at least about 85%, 90%, 95%,
99% or more identical o, and/or capable of hybridizing under the stringency conditions
described herein).
In an embodiment, the nucleic acid further comprises a nucleotide sequence encoding at least
one, two, or three CDRs from a heavy chain variable region of a polypeptide (e.g., an antibody
molecule or TCR le) described herein, or a nucleotide sequence substantially homologous
thereto (e.g., a sequence at least about 85%, 90%, 95%, 99% or more identical thereto, and/or e
of hybridizing under the stringency conditions described herein). In r embodiment, the nucleic
acid further comprises a nucleotide sequence ng at least one, two, or three CDRs from a light
chain variable region of a polypeptide (e.g., an antibody molecule or TCR molecule) described herein,
or a nucleotide sequence substantially homologous o (e.g., a ce at least about 85%, 90%,
95%, 99% or more identical thereto, and/or capable of hybridizing under the stringency conditions
bed herein). In yet r embodiment, the nucleic acid comprises a tide ce
encoding at least one, two, three, four, five, or six CDRs from heavy and light chain variable regions
of a polypeptide (e.g., an antibody molecule or TCR molecule) described herein, or a nucleotide
sequence substantially homologous thereto (e.g., a sequence at least about 85%, 90%, 95%, 99% or
more identical thereto, and/or capable of hybridizing under the ency conditions described
herein) .
2017/068204
In an embodiment, the nucleic acid comprises a portion of a nucleotide sequence described
herein. The portion may encode, for example, a variable region (e.g., VH or VL); one, two, or three
or more (e.g., four, five, or six) CDRs; or one, two, three, or four or more framework s,
ally, a constant region or an Fc region.
The nucleic acids disclosed herein include deoxyribonucleotides or ribonucleotides, or
s thereof. The polynucleotide may be either single-stranded or double-stranded, and if single-
stranded may be the coding strand or non-coding (antisense) strand. A polynucleotide may comprise
d nucleotides, such as methylated nucleotides and nucleotide analogs. The sequence of
nucleotides may be interrupted by non-nucleotide components. A polynucleotide may be further
modified after polymerization, such as by conjugation with a labeling component. The nucleic acid
may be a recombinant polynucleotide, or a polynucleotide of genomic, cDNA, semisynthetic, or
tic origin which either does not occur in nature or is linked to another polynucleotide in a non-
natural arrangement.
In some aspects, the application features host cells and vectors containing the c acids
described herein. The c acids may be present in a single vector or separate vectors present in
the same host cell or separate host cell, as described in more detail below.
Vectors
The present disclosure features vectors that comprise nucleotide sequences encoding
polypeptides (e.g., binding polypeptides, e.g., antibody les or TCR molecules).
The vectors include, but are not limited to, a virus, plasmid, cosmid, lambda phage or a yeast
artificial chromosome (YAC).
Numerous vector systems can be ed. For example, one class of vectors utilizes DNA
elements which are d from animal viruses such as, for example, bovine papilloma virus,
polyoma Virus, adenovirus, vaccinia virus, baculovirus, retroviruses (Rous Sarcoma Virus, MMTV or
MOMLV) or SV4O virus. Another class of vectors utilizes RNA elements derived from RNA viruses
such as Semliki Forest Virus, Eastern Equine Encephalitis Virus and Flaviviruses.
Additionally, cells which have stably ated the DNA into their chromosomes may be
selected by introducing one or more markers which allow for the selection of transfected host cells.
The marker may provide, for example, prototropy to an ophic host, biocide resistance (e.g.,
antibiotics), or resistance to heavy metals such as copper, or the like. The selectable marker gene can
be either directly linked to the DNA ces to be sed, or introduced into the same cell by
cotransformation. Additional elements may also be needed for optimal synthesis of mRNA. These
elements may include splice signals, as well as transcriptional promoters, enhancers, and termination
signals.
Once the expression vector or DNA ce containing the constructs has been prepared for
expression, the expression vectors may be transfected or introduced into an appropriate host cell.
Various techniques may be employed to e this, such as, for example, protoplast fusion, calcium
phosphate precipitation, electroporation, retroviral transduction, viral transfection, gene gun, lipid
based transfection or other conventional techniques. In the case of protoplast fusion, the cells are
grown in media and screened for the appropriate activity.
Methods and conditions for culturing the resulting transfected cells and for recovering the
polypeptide (e. g., antibody molecule) produced are known to those skilled in the art, and may be
varied or optimized depending upon the specific expression vector and mammalian host cell
ed, based upon the present description.
Cells
The present sure also provides host cells comprising a nucleic acid encoding a
polypeptide (e.g., an antibody molecule or TCR molecule) as described herein. The ptide (e.g.,
antibody molecule or TCR molecule) can be engineered in accordance with a method described
herein. For example, the host cells may comprise a nucleic acid molecule having a nucleotide
sequence of a polypeptide described herein (e.g., an dy molecule or TCR molecule described
herein), a ce ntially homologous thereto (e.g., a sequence at least about 85%, 90%, 95%,
99% or more identical thereto, and/or capable of hybridizing under the stringency conditions
described ), or a portion of one of said nucleic acids.
In some embodiments, the host cells are genetically engineered to comprise nucleic acids
encoding the polypeptide (e.g., antibody molecule or TCR molecule) described herein.
In certain embodiments, the host cells are genetically engineered by using an expression
cassette. The phrase ssion cassette,” refers to nucleotide sequences, which are capable of
affecting expression of a gene in hosts compatible with such sequences. Such cassettes may e a
promoter, an open reading frame with or without introns, and a termination signal. Additional factors
necessary or helpful in effecting expression may also be used, such as, for example, an ble
promoter.
The disclosure also provides host cells comprising the vectors described herein.
The cell can be, but is not limited to, a eukaryotic cell, a bacterial cell, an insect cell, or a
human cell. Suitable eukaryotic cells include, but are not limited to, Vero cells, HeLa cells, COS cells,
CHO cells, HEK293 cells, BHK cells and MDCKII cells. Suitable insect cells include, but are not
limited to, Sf9 cells.
Uses of Polypeptides
The polypeptides (e.g., binding polypeptides, e.g., dy les or TCR molecule)
disclosed herein, as well as the ceutical compositions sed herein, have in vitro, ex vivo,
and in viva therapeutic, prophylactic, and/or diagnostic utilities.
In an embodiment, the ptide (e.g., antibody molecule or TCR le) modulates
(e.g., reduces (e.g., inhibits, blocks, or neutralizes) or increases (e.g., activates, initiates, or es))
one or more ical activities of a target molecule (e. g., protein) or cell. For example, these
polypeptides (e.g., antibody molecules or TCR les) can be administered to cells in culture, in
vitra or ex viva, or to a subject, e.g., a human subject, e.g., in viva, to modulate one or more biological
activities of the target molecule or cell. Accordingly, in an aspect, the disclosure provides a method
of ng, ting, or diagnosing a disorder, e. g., a er described herein, in a subject,
comprising administering to the subject a polypeptide (e.g., an antibody molecule or TCR molecule)
described herein, such that the disorder is treated, prevented, or diagnosed. For e, the
disclosure provides a method comprising contacting the polypeptide (e.g., dy molecule or TCR
molecule) described herein with cells in culture, 6.3. in vitra or ex viva, or administering the
polypeptide (e.g., antibody molecule or TCR molecule) described herein to a subject, e.g., in viva, to
treat, prevent, or se a disorder, e.g., a disorder ated with a target molecule or cell (e.g., a
disorder described herein).
As used herein, the term “subject” is intended to include human and non-human animals. In
some embodiments, the subject is a human subject, e.g., a human patient having a disorder described
herein, or at risk of having a disorder described herein. The term uman animals” includes
mammals and non-mammals, such as non-human primates. In an embodiment, the subject is a
human. The methods and compositions described herein are suitable for treating human patients for a
disorder described herein. ts having a disorder bed herein include those who have
developed a disorder described herein but are (at least temporarily) asymptomatic, patients who have
exhibited a symptom of a disorder described herein, or patients having a disorder related to or
associated with a disorder described herein.
Methods of Treating or Preventing Disorders
The polypeptides (e.g., antibody molecules or TCR les) described herein can be used
to treat or prevent disorders or conditions. In an embodiment, the polypeptide has an optimal or
improved half-life, which can be desirable for treating or preventing the disorder or condition. While
not wishing to be bound by theory, it is believed that in an embodiment, the polypeptide bed
herein (e.g., the polypeptide having an optimal or improved half-life) can provide one or more
benefits over another polypeptide having the same or similar binding affinity and/or specificity (e.g., a
polypeptide that does not have, or has not been engineered to have, an optimal or improved half-life).
These benefits can include, but are not d to, an increased therapeutic or preventive efficacy, a
reduced dosage regimen, or an improved pharmacokinetic property. In an ment, the
polypeptide includes a mutated Fc region as described herein.
Exemplary disorders or conditions that can be d or ted by the polypeptides
described herein include, but are not limited to, a cancer (e.g., a solid tumor or a hematologic cancer),
an infectious disease (e.g., a bacterial infection or a viral infection), an immune disorder (e.g., an
autoimmune er), an inflammatory disorder, a lic disorder (e.g., diabetes), a
cardiovascular disorder, an organ transplant rejection.
Exemplary cancers that can be d or prevented by the polypeptides described herein
include, but are not limited to, acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML),
adrenocortical carcinoma, Kaposi sarcoma, an AIDS -related lymphoma, primary central nervous
system (CNS) ma, anal cancer, appendix cancer, astrocytoma, atypical teratoid/rhabdoid
tumor, basal cell carcinoma, bile duct cancer, bladder cancer, bone cancer (e.g., Ewing sarcoma or
osteosarcoma and malignant fibrous histiocytoma), brain tumor (e.g., astrocytomas, brain stem
glioma, central nervous system atypical teratoid/rhabdoid tumor, central nervous system embryonal
tumor, central nervous system germ cell tumor, craniopharyngioma, or ependymoma), breast cancer,
bronchial tumor, Burkitt lymphoma, carcinoid tumor (e.g., gastrointestinal carcinoid tumor), cardiac
(heart) tumor, embryonal tumor, germ cell tumor, lymphoma, cervical , giocarcinoma,
chordoma, chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia (CML), chronic
myeloproliferative neoplasm, colon cancer, colorectal cancer, craniopharyngioma, cutaneous T-cell
lymphoma, ductal carcinoma in situ (DCIS), endometrial cancer, moma, esophageal cancer,
esthesioneuroblastoma, Ewing sarcoma, extracranial germ cell tumor, extragonadal germ cell tumor,
eye cancer (e.g., intraocular melanoma or retinoblastoma), fallopian tube cancer, fibrous histiocytoma
of bone, osteosarcoma, gallbladder cancer, gastric ch) cancer, gastrointestinal carcinoid tumor,
gastrointestinal stromal tumors (GIST), germ cell tumor (e. g., central nervous system tumor,
ranial tumor, onadal tumor, ovarian cancer, or testicular ), ional trophoblastic
disease, glioma, hairy cell leukemia, head and neck cancer, cellular (liver) , Hodgkin
ma, hypopharyngeal , intraocular melanoma, islet cell tumor, pancreatic neuroendocrine
tumor, Kaposi sarcoma, kidney cancer (e.g., renal cell cancer or Wilms tumor), Langerhans cell
histiocytosis (LCH), laryngeal cancer, leukemia (e.g., acute lymphoblastic leukemia (ALL), acute
myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia
(CML), or hairy cell leukemia), lip and oral cavity cancer, liver cancer, lung cancer (e.g., non-small
cell lung cancer (NSCLC) or small cell lung cancer), lymphoma (e.g., aids-related, Burkitt lymphoma,
cutaneous T-cell lymphoma, Hodgkin lymphoma, non-Hodgkin lymphoma, or primary central
nervous system (CNS) lymphoma), Waldenstrom macroglobulinemia, male breast cancer, malignant
fibrous histiocytoma of bone and osteosarcoma, melanoma (e.g., intraocular (eye) melanoma), Merkel
cell oma, mesothelioma, metastatic squamous neck cancer, midline tract carcinoma, mouth
cancer, multiple endocrine neoplasia syndrome, multiple myeloma/plasma cell sm, mycosis
fungoides, myelodysplastic syndrome, myelodysplastic/myeloproliferative sm, chronic
myeloproliferative neoplasm, nasal cavity and paranasal sinus , nasopharyngeal cancer,
neuroblastoma, oral cancer, lip and oral cavity cancer, oropharyngeal cancer, osteosarcoma and
malignant fibrous cytoma of bone, ovarian cancer (e.g., lial ovarian cancer or germ cell
ovarian tumor), pancreatic cancer, pancreatic neuroendocrine tumors (islet cell tumors),
papillomatosis, paraganglioma, paranasal sinus and nasal cavity cancer, parathyroid cancer, penile
cancer, pharyngeal cancer, pheochromocytoma, pituitary tumor, pleuropulmonary blastoma,
peritoneal , prostate cancer, rectal cancer, retinoblastoma, rhabdomyosarcoma, ry gland
, a (e. g., Ewing a, Kaposi sarcoma, osteosarcoma, rhabdomyosarcoma, soft tissue
sarcoma, or e sarcoma), Sézary me, skin cancer (e.g., melanoma, Merkel cell carcinoma,
or nonmelanoma skin cancer), small ine cancer, squamous cell carcinoma, testicular cancer,
throat cancer, thymoma and thymic carcinoma, thyroid cancer, transitional cell cancer of the renal
pelvis and ureter, urethral cancer, endometrial uterine cancer, vaginal cancer, vulvar cancer, or a
atic lesion thereof.
Exemplary infectious diseases that can be treated or prevented by the ptides described
herein e, but are not limited to, Acinetobacter infections, actinomycosis, African ng
sickness (African trypanosomiasis), AIDS (acquired immunodeficiency syndrome), amebiasis,
anaplasmosis, angiostrongyliasis, anisakiasis, anthrax, arcanobacterium haemolyticum ion,
argentine hemorrhagic fever, ascariasis, aspergillosis, irus infection, babesiosis, bacillus
cereus ion, bacterial pneumonia, bacterial vaginosis, bacteroides infection, balantidiasis,
bartonellosis, baylisascaris infection, bk virus infection, black , blastocystosis, blastomycosis,
bolivian hemorrhagic fever, botulism (and infant botulism), brazilian hemorrhagic fever, brucellosis,
bubonic plague, burkholderia infection, buruli ulcer, calicivirus infection (norovirus and sapovirus),
campylobacteriosis, candidiasis (moniliasis; thrush), capillariasis, carrion’s disease, cat-scratch
disease, cellulitis, chagas disease (american trypanosomiasis), chancroid, chickenpox, chikungunya,
chlamydia, chlamydophila pneumoniae infection (taiwan acute respiratory agent or twar), cholera,
blastomycosis, chytridiomycosis, clonorchiasis, clostridium difficile colitis,
coccidioidomycosis, do tick fever (CTF), common cold (Acute viral haryngitis; Acute
coryza), Creutzfeldt-Jakob disease (CJD), Crimean-Congo hemorrhagic fever (CCHF),
cryptococcosis, cryptosporidiosis, cutaneous larva migrans (CLM), cyclosporiasis, cysticercosis,
cytomegalovirus infection, dengue fever, desmodesmus infection, dientamoebiasis, diphtheria,
diphyllobothriasis, dracunculiasis, ebola hemorrhagic fever, echinococcosis, ehrlichiosis,
enterobiasis (pinworm infection), enterococcus infection, enterovirus infection, epidemic typhus,
ma infectiosum (fifth disease), exanthem subitum (sixth disease), fasciolasis, fasciolopsiasis,
fatal al insomnia (FFI), filariasis, food poisoning by clostridium perfringens, free-living amebic
infection, fusobacterium infection, gas gangrene (clostridial rosis), geotrichosis, gerstmann-
straussler-scheinker syndrome (GSS), giardiasis, rs, gnathostomiasis, gonorrhea, granuloma
inguinale (donovanosis), Group A streptococcal infection, Group B streptococcal infection,
hilus influenzae infection, hand, foot and mouth disease (HFMD), Hantavirus Pulmonary
Syndrome (HPS), and virus disease, helicobacter pylori infection, hemolytic-uremic
syndrome (HUS), hemorrhagic fever with renal syndrome (HFRS), hepatitis A, hepatitis B, hepatitis
C, hepatitis D, hepatitis E, herpes simplex, histoplasmosis, hookworm infection, human
bocavirus infection, human ewingii hiosis, human granulocytic anaplasmosis (HGA), human
metapneumovirus infection, Human monocytic ehrlichiosis, human papillomavirus (HPV) ion,
Human parainfluenza virus infection, Hymenolepiasis, Epstein-Barr Virus Infectious
Mononucleosis (Mono), za (flu), isosporiasis, kawasaki disease, keratitis, kingella
kingae infection, kuru, lassa fever, legionellosis (legionnaires' disease), legionellosis (pontiac fever),
leishmaniasis, leprosy, leptospirosis, iosis, lyme disease (lyme borreliosis), lymphatic
sis (Elephantiasis), Lymphocytic choriomeningitis, Malaria, Marng hagic
fever (MHF), Measles, Middle East respiratory syndrome (MERS), melioidosis (Whitmore's e),
meningitis, ococcal disease, metagonimiasis, microsporidiosis, molluscum contagiosum (MC),
Monkeypox, Mumps, Murine typhus (Endemic typhus), Mycoplasma pneumonia, ma
(disambiguation), Myiasis, Neonatal conjunctivitis (Ophthalmia neonatorum), (New) Variant
Creutzfeldt-Jakob disease (vCJD, nvCJD), nocardiosis, onchocerciasis (River blindness),
opisthorchiasis, paracoccidioidomycosis (South American blastomycosis), paragonimiasis,
rellosis, pediculosis capitis (head lice), pediculosis corporis (body lice), pediculosis
pubis (pubic lice, crab lice), pelvic inflammatory e (PID), pertussis (Whooping cough), plague,
pneumococcal ion, pneumocystis pneumonia (PCP), pneumonia, yelitis,
prevotella infection, primary amoebic meningoencephalitis (PAM), progressive multifocal
leukoencephalopathy, psittacosis, Q fever, rabies, relapsing fever, respiratory syncytial
virus infection, rhinosporidiosis, rhinovirus infection, tsial infection, rickettsialpox, Rift Valley
fever (RVF), Rocky Mountain spotted fever (RMSF), rotavirus infection, rubella, salmonellosis,
SARS e Acute Respiratory Syndrome), scabies, schistosomiasis, sepsis, shigellosis (Bacillary
dysentery), shingles s zoster), smallpox (Variola), sporotrichosis, staphylococcal food
poisoning, staphylococcal infection, strongyloidiasis, subacute sclerosing ephalitis, syphilis,
Taeniasis, Tetanus (Lockj aw), Tinea barbae (Barber's itch), Tinea capitis (Ringworm of the Scalp),
Tinea corporis (Ringworm of the Body), Tinea cruris (Jock itch), Tinea manum (Ringworm of the
Hand), Tinea nigra, Tinea pedis (Athlete’s foot), Tinea unguium omycosis), Tinea
versicolor (Pityriasis olor), Toxocariasis r Larva Migrans (OLM)), Toxocariasis (Visceral
Larva Migrans (VLM)), Trachoma, Toxoplasmosis, Trichinosis, Trichomoniasis,
Trichuriasis (Whipworm infection), Tuberculosis, Tularemia, Typhoid fever, Typhus fever,
Ureaplasma urealyticum infection, Valley fever, Venezuelan equine encephalitis, Venezuelan
hemorrhagic fever, Vibrio vulnificus infection, Vibrio parahaemolyticus enteritis, viral pneumonia,
West Nile Fever, white piedra (Tinea blanca), Yersinia pseudotuberculosis infection, yersiniosis,
yellow fever, Zika fever, or cosis.
Exemplary immune disorders or conditions that can be treated or prevented by the
polypeptides described herein include, but are not limited to, Addison’s disease,
agammaglobulinemia, alopecia areata, amyloidosis, ankylosing litis, anti-GBM/anti-TBM
tis, antiphospholipid syndrome (APS), autoimmune hepatitis, mune inner ear disease
(AIED), axonal & neuronal athy (AMAN), Behcet’s e, Bullous pemphigoid, Castleman
e (CD), Celiac disease, Chagas disease, chronic inflammatory demyelinating polyneuropathy
(CIDP), chronic recurrent multifocal osteomyelitis (CRMO), Churg-Strauss, Cicatricial
pemphigoidfbenign mucosal pemphigoid, Cogan’s syndrome, Cold agglutinin disease, Congenital
heart block, kie myocarditis, CREST syndrome, Crohn’ s disease, dermatitis herpetiformis,
dermatomyositis, Devic’s disease (neuromyelitis optica), Discoid lupus, Dressler’s syndrome,
endometriosis, eosinophilic esophagitis (EoE), philic fasciitis, erythema nodosum, essential
mixed cryoglobulinemia, Evans syndrome, fibromyalgia, fibrosing alveolitis, giant cell arteritis
(temporal arteritis), giant cell myocarditis, Glomerulonephritis, sture’s syndrome,
Granulomatosis with Polyangiitis, Graves’ e, Guillain-Barre me, Hashimoto’s thyroiditis,
hemolytic anemia, Henoch-Schonlein purpura (HSP), herpes gestationis or pemphigoid gestationis
(PG), mmalglobulinemia, IgA nephropathy, IgG4-related sing disease, inclusion body
myositis (IBM), titial cystitis (IC), juvenile arthritis, juvenile diabetes (Type 1 diabetes),
juvenile myositis (JM), Kawasaki disease, Lambert-Eaton syndrome, leukocytoclastic vasculitis,
Lichen planus, Lichen sclerosus, us conjunctivitis, linear IgA disease (LAD), lupus, Lyme
disease chronic, Meniere’s disease, microscopic polyangiitis (MPA), mixed tive tissue disease
(MCTD), Mooren’s ulcer, Habermann disease, multiple sclerosis (MS), Myasthenia gravis,
Myositis, Narcolepsy, Neuromyelitis optica, neutropenia, ocular cicatricial pemphigoid, optic neuritis,
palindromic rheumatism (PR), PANDAS (Pediatric Autoimmune Neuropsychiatric Disorders
Associated with Streptococcus), paraneoplastic cerebellar degeneration (PCD), Paroxysmal nocturnal
obinuria (PNH), Parry Romberg syndrome, Pars is (peripheral uveitis), Parsonnage-
Turner syndrome, Pemphigus, eral neuropathy, Perivenous encephalomyelitis, pernicious
anemia (PA), POEMS syndrome (polyneuropathy, megaly, endocrinopathy, monoclonal
athy, skin changes), polyarteritis nodosa, polymyalgia rheumatica, polymyositis,
postmyocardial infarction syndrome, postpericardiotomy me, primary biliary cirrhosis, primary
sclerosing cholangitis, progesterone itis, psoriasis, psoriatic arthritis, pure red cell aplasia
(PRCA), pyoderma gangrenosum, Raynaud’s phenomenon, Reactive Arthritis, Reflex sympathetic
dystrophy, Reiter’s syndrome, relapsing polychondritis, restless legs syndrome (RLS), retroperitoneal
fibrosis, rheumatic fever, rheumatoid arthritis (RA), sarcoidosis, Schmidt syndrome, scleritis,
scleroderma, Sj ogren’s syndrome, sperm & testicular munity, Stiff person syndrome (SPS),
subacute bacterial endocarditis (SBE), Susac’s syndrome, sympathetic ophthalmia (SO), Takayasu’s
arteritis, temporal arteritis/Giant cell arteritis, thrombocytopenic purpura (TTP), Tolosa-Hunt
syndrome (THS), transverse myelitis, type 1 diabetes, ulcerative colitis (UC), erentiated
tive tissue disease (UCTD), uveitis, vasculitis, vitiligo, or Wegener’s granulomatosis
(Granulomatosis with Polyangiitis (GPA)).
The polypeptides (e.g., antibody molecules or TCR molecules) described herein are typically
administered at a frequency that keeps a therapeutically effective level of polypeptides in the patient’s
system until the patient recovers. For example, the polypeptides may be administered at a frequency
that achieves a serum concentration ient for at least about 1, 2, 5, 10, 20, 30, or 40 polypeptides
to bind each target molecule or cell. In an embodiment, the polypeptides are administered every 1, 2,
3, 4, 5,6, or 7 days, every 1,2,3, 4, 5, or 6 weeks, or every 1, 2, 3, 4,5, or 6 months.
Methods of administering various polypeptides (e.g., antibody molecules or TCR molecules)
are known in the art and are described below. le s of the polypeptides used will depend
on the age and weight of the subject and the particular drug used.
The polypeptides can be used by themselves or conjugated to a second agent, e.g., an bacterial
agent, toxin, or protein, e.g., a second polypeptide. This method includes: administering the
polypeptide, alone or conjugated to a second agent, to a subject requiring such ent. The
ptides can be used to deliver a variety of therapeutic agents, e.g., a toxin, or mixtures thereof.
Combination Therapies
The polypeptides (e.g., antibody molecules or TCR molecules) can be used in combination
with other therapies. For example, the combination y can include a ptide mulated
with, and/or co-administered with, one or more additional therapeutic , e. 3., one or more
additional therapeutic agents described herein. In other embodiments, the polypeptides are
administered in combination with other therapeutic treatment modalities, e.g., other therapeutic
treatment modalities described herein. Such combination ies may advantageously utilize lower
dosages of the administered therapeutic agents, thus avoiding possible toxicities or complications
associated with the various monotherapies.
stered “in combination”, as used herein, means that two (or more) different treatments
are delivered to the subject before, or during the course of the subject's affliction with a disorder. In
an embodiment, two or more treatments are red prophylactically, e.g., before the subject has the
disorder or is diagnosed with the disorder. In another embodiment, the two or more treatments are
delivered after the subject has developed or diagnosed with the disorder. In some embodiments, the
delivery of one treatment is still occurring when the delivery of the second begins, so that there is
overlap. This is sometimes referred to herein as "simultaneous" or "concurrent delivery." In other
embodiments, the delivery of one treatment ends before the delivery of the other treatment begins. In
some embodiments of either case, the treatment is more effective because of ed administration.
For example, the second treatment is more effective, e.g., an equivalent effect is seen with less of the
second treatment, or the second treatment reduces symptoms to a greater extent, than would be seen if
the second treatment were administered in the absence of the first treatment, or the analogous
situation is seen with the first ent. In some embodiments, delivery is such that the reduction in
a m, or other parameter related to the disorder is greater than what would be observed with one
treatment delivered in the absence of the other. The effect of the two treatments can be partially
additive, wholly additive, or greater than additive. The delivery can be such that an effect of the first
treatment delivered is still detectable when the second is delivered.
In an embodiment, the polypeptide is administered in combination with a second therapy
(e. g., an additional agent) to treat or t a er described herein. In an embodiment, the
additional agent is a second polypeptide (e.g., antibody molecule), e.g., a polypeptide (e.g., an
antibody molecule) different from a first ptide (e.g., antibody molecule). Exemplary
polypeptides (e.g., antibody molecules) that can be used in combination include, but are not limited
to, any combination of the polypeptides (e.g., antibody molecules) described herein. In another
embodiment, the additional agent is other than a polypeptide (e.g., antibody le). For example,
the additional agent can be a small molecule or a c acid molecule. In yet r embodiment,
the second therapy is chosen from a surgery, a radiation therapy, a cell therapy (e.g., a stem cell
therapy), or an organ or tissue transplantation.
In an embodiment, the second therapy comprises a therapy chosen from one or more of: an
androgen replacement therapy, an antihormone therapy, an antiserum y, an autologous immune
enhancement therapy, a biotherapy, a blood irradiation y, a brachytherapy, a cardiac
resynchronization therapy, a cell therapy, a cell transfer therapy, a chelation therapy, a chemotherapy,
a chrysotherapy, a cobalt therapy, a cold compression therapy, a cryotherapy, an electroconvulsive
y, an electromagnetic therapy, an electron therapy, an electrotherapy, an enzyme ement
therapy, an epigenetic therapy, an estrogen replacement therapy, an orporeal shockwave
therapy, a fast neutron therapy, a fluoride therapy, a gene therapy, a heat therapy, a helminthic
therapy, a hormone y, a hormone ement therapy, a host modulatory therapy, a aric
oxygen therapy, a hyperthermia therapy, an immunosuppressive therapy, an immunotherapy, an
perative electron radiation y, an intraoperative radiation therapy, an inversion therapy, a
laser therapy, a light therapy, a lithium therapy, a low level laser therapy, a magnet therapy, a
magnetic resonance therapy, a medical gas therapy, a medical nutrition therapy, a molecular
chaperone therapy, a molecular therapy, a monoclonal dy therapy, a negative air tion
therapy, a neutron e therapy, a neutron therapy, an oral rehydration therapy, an osmotherapy, an
oxygen therapy, an ozone therapy, a palliative therapy, a particle therapy, a phage therapy, a
phonemic neurological hypochromium therapy, a photodynamic therapy, a phototherapy, a
photothermal therapy, a physical y, a prolotherapy, a protein therapy, a proton therapy, a pulsed
electromagnetic field therapy, a PUVA therapy, a radiation therapy, a rehydration therapy, a
respiratory y, salvage therapy, a serotherapy, a stem cell therapy, a stereotactic radiation
therapy, a targeted therapy, a thermotherapy, a TK cell therapy, a tolerogenic y, a transdermal
continuous oxygen therapy, an ultraviolet light therapy, or a virotherapy.
Exemplary therapies that can be used in ation with a polypeptide or composition
described herein to treat or prevent other disorders are also described in the section of “Methods of
Treating or Preventing Disorders” herein.
Methods of Diagnosis
In some aspects, the present disclosure provides a diagnostic method for detecting the
presence of a target le (e.g., a protein) or cell in vitro (e.g., in a biological sample, such as a
biopsy or body fluid (e.g., blood, urine, or cerebrospinal fluid) sample) or in viva (e.g., in vivo
imaging in a subject). The method includes: (i) contacting the sample with a polypeptide described
herein (e.g., an antibody molecule described herein), or administering to the subject, the polypeptide
(6.3., antibody le); (optionally) (ii) contacting a reference sample, e.g., a control sample (e.g., a
control biological sample, such as a biopsy or body fluid (e.g., blood, urine, or cerebrospinal fluid)
) or a control subject with a ptide described herein (e.g., an antibody molecule described
herein); and (iii) detecting formation of a complex between the polypeptide (e. g., antibody molecule)
and the target molecule or cell in the sample or subject, or the control sample or subject, wherein a
change, e.g., a statistically significant change, in the formation of the x in the sample or subject
relative to the control sample or subject is indicative of the ce of the target molecule or cell in
the sample. The polypeptide (e.g., antibody molecule) can be directly or indirectly labeled with a
detectable substance to facilitate detection of the bound or d polypeptide (e.g., dy
molecule). Suitable detectable substances include various enzymes, prosthetic groups, fluorescent
materials, luminescent als and radioactive materials, as described herein.
The term “sample,” as it refers to samples used for detecting bacteria includes, but is not
limited to, cells, cell lysates, proteins or ne ts of cells, body fluids such as blood, urine,
or CSF, or tissue samples such as biopsies.
Complex formation between the polypeptide (e.g., dy molecule), and the target
molecule or cell, can be detected by measuring or visualizing either the polypeptide (e.g., antibody
molecule) bound to the target molecule or cell, or unbound polypeptide (e.g., antibody molecule).
Any suitable detection assays can be used, and conventional ion assays include an enzyme-
linked imrnunosorbent assay (ELISA), a radioimmunoassay (RIA), a FACS assay, a BIACORE assay,
or tissue immunohistochemistry. Alternative to labeling the ptide, the ce of the target
molecule or cell can be assayed in a sample by a ition immunoassay utilizing standards labeled
with a detectable substance and an unlabeled polypeptide. In this assay, the biological sample, the
labeled standards and the polypeptide are combined and the amount of labeled standard bound to the
unlabeled g molecule is determined. The amount of the target molecule or cell in the sample is
inversely proportional to the amount of labeled standard bound to the polypeptide (e.g., antibody
molecule).
The polypeptides (e.g., antibody molecules) described herein can be used to diagnose
disorders that can be d or ted by the polypeptides described herein. The detection or
diagnostic methods described herein can be used in combination with other methods described herein
to treat or prevent ers described herein.
Additional embodiments are described in the numbered paragraphs below.
1. A method of making a nucleic acid sequence comprising a sequence that encodes a heavy
chain element (HC element) of an antibody heavy chain variable region (HCVR) and a light chain
element (LC element) of an antibody light chain variable region (LCVR), and wherein the HCVR and
LCVR are matched, the method comprising:
a) acquiring an isolated production reaction site, e.g., a production micro-chamber,
sing:
i) a heavy chain (HC) strand, wherein the HC strand is a strand of a heavy chain
double-stranded cDNA (HC ds cDNA) comprising a segment that encodes an HC element of
the HCVR from a cell, e.g., a heavy chain variable region sequence (HCVRS); and
ii) a light chain (LC) , wherein the LC strand is a strand of a light chain double-
stranded cDNA (LC ds cDNA) comprising a segment that encodes an LC element of the
LCVR from the cell, e. 3., a light chain variable region sequence (LCVRS), and
b) covalent linking, e.g., ligation, of an HC strand to an LC strand,
wherein the isolated production on site, e.g., a production micro-chamber, does not
include a nucleic acid encoding an LCVR or an HCVR from a cell other than the cell,
thereby making a c acid sequence comprising a sequence that encodes an HC element
of an HCVR and a LC element of an LCVR, wherein the HCVR and LCVR are matched.
2. The method of paragraph 1, wherein the HC element comprises, or consists of, an
HCVRS, or a functional fragment thereof (e.g., an antigen binding fragment thereof).
3. The method of paragraph 1 or 2, n the LC element comprises, or consists of, an
LCVRS, or a functional fragment thereof (e.g., an antigen binding fragment thereof).
4. The method of any of paragraphs 1-3, wherein the nucleic acid sequence is configured
such that, when expressed, the HCVRS and the LCVRS form a functional antigen binding molecule,
e.g., an scFv.
. The method of aph 4, wherein the n binding molecule, e.g., an scFv, is
functional in vitro, ex vivo, or in viva, e.g., as determined by a method or assay described herein.
6. The method of any of aphs 1-5, n acquiring an ed production reaction
site, e.g., a production micro-chamber, comprises:
a) acquiring a capture ate bound to:
(i) a first double-stranded cDNA (ds cDNA) comprising a strand that is
complementary to a first mRNA that encodes an HCVR from a cell; and
2017/068204
(ii) a second ds cDNA comprising a strand complementary to a second mRNA
encoding an LCVR from the cell (the cDNA loaded capture substrate), and
b) maintaining the isolated production reaction site, e.g., the production chamber,
under conditions that allow amplification of the first and second ds cDNAs, to produce:
a plurality of HC ds cDNAs comprising a segment that encodes an HC element of the HCVR
from the cell, e.g., an HCVRS; and
a plurality of LC ds cDNAs sing a segment that encodes an LC element of the LCVR
from the cell, e.g., an LCVRS.
7. The method of paragraph 6, wherein the HC ds cDNA is identical, or substantially
identical, to the first ds cDNA.
8. The method of paragraph 6 or 7, wherein the LC ds cDNA is cal, or substantially
identical, to the second ds cDNA.
9. The method of any of paragraphs 6-8, wherein the capture substrate comprises a bead, e. g.,
a magnetic bead.
10. The method of any of paragraphs 6-9, wherein the capture ate comprises a moiety
(e.g., an oligonucleotide) which binds to cDNA, e.g., (i) a moiety which binds to the HC strand; (ii) a
moiety which binds to the LC strand; or (iii) both (i) and (ii).
11. The method of any of paragraphs 6-10, wherein the first mRNA and the second mRNA
are disposed on an mRNA loaded capture substrate.
12. The method of any of paragraphs 6-11, wherein the isolated tion on site, e.g.,
the production micro-chamber, comprises:
a t mixture suitable for ing, from the first and second mRNAs (e. g., after the
first and second mRNAs are released from the mRNA loaded capture substrate into a solution), a first
ds cDNA sing a segment that encodes an HC element of the HCVR of the cell, e. g., an
HCVRS, and a second ds cDNA comprising a t that encodes an LC element of the LCVR of
the cell, e.g., an LCVRS.
13. The method of any of paragraphs 6-12, wherein the isolated production reaction site, e.g.,
production micro-chamber, comprises primers that e the production of the first ds cDNA.
14. The method of any of paragraphs 6-13, wherein the isolated production reaction site, e.g.,
production micro-chamber, comprises primers that mediate the production of the second ds cDNA.
. The method of any of paragraphs 6-14, wherein a cDNA strand that is complementary to
a first mRNA that encodes an HCVR from a cell is made by reverse transcription of the first mRNA.
16. The method of any of paragraphs 6-15, wherein a cDNA strand that is complementary to
a second mRNA that encodes an LCVR from a cell is made by reverse transcription of the second
mRNA.
17. The method of paragraph 15 or 16, wherein the reverse transcription takes place in the
isolated production reaction site, e.g., a production-micro chamber.
18. The method of paragraph 15 or 16, wherein the reverse transcription takes place in an
isolated cell reaction site, e.g., a cell isolation micro-chamber.
19. The method of aph 15 or 16, wherein the reverse transcription takes place outside
the isolated production reaction site, e.g., a production micro-chamber, or outside an isolated cell
reaction site, e. g., a cell isolation micro-chamber.
. The method of paragraph 15 or 16, wherein the reverse transcription takes place outside
the isolated production reaction site, e. g., a production-micro chamber, and outside an ed cell
on site, e.g., a cell isolation micro-chamber.
21. The method of aph 15 or 16, wherein the reverse transcription takes place outside
an isolated on site, e.g., outside a micro-chamber.
22. The method of any of paragraphs 6-21, wherein the amplification comprises 20 or fewer
cycles, e.g., 15 or fewer, 14 or fewer, 13 or fewer, 12 or fewer, 11 or fewer, 10 or fewer, 9 or fewer, 8
or fewer, 7 or fewer, 6 or fewer, or 5 or fewer cycles.
23. The method of any of aphs 6-22, wherein the reverse transcription and/or
amplification uses one or more primers, e. g., comprising a sequence specific for an HCVRS and/or an
LCVRS.
24. The method of any of paragraphs 6-23, wherein the amplification comprises using two or
more primers that e the production of the HC ds cDNA, wherein at least one primer comprises
a nucleotide ation, and wherein at least one primer does not comprise a nucleotide
modification.
. The method of any of paragraphs 6-24, wherein the amplification comprises using two or
more primers that mediate the production of the LC ds cDNA, wherein at least one primer comprises
a nucleotide modification, and wherein at least one primer does not comprise a nucleotide
ation.
26. The method of paragraph 25, wherein at least one primer comprises a nucleotide
modification, e.g., which reduces, e.g., inhibits, DNA synthesis, e.g., by a DNA rase.
27. The method of paragraph 25 or 26, wherein at least one primer does not comprise a
nucleotide modification, e.g., which reduces, e.g., inhibits, DNA synthesis, e.g., by a DNA
polymerase.
28. The method of paragraph 26 or 27, wherein the nucleotide modification inhibits a DNA
polymerase from extending the DNA.
29. The method of any of paragraphs 26-28, wherein the nucleotide modification is an
insertion of a spacer to the primer, e.g., between two adjacent nucleotides in the .
. The method of aph 29, wherein the spacer is a flexible , a carbon spacer
(e.g., -(CH2)n-, wherein n=3, 4, 5, or more), two or more (e.g., three, four, five, or more) abasic
nucleotides or a PEG spacer.
WO 19402 2017/068204
31. The method of any of paragraphs 26-28, n the nucleotide modification is 2’-O-
methyl, 2’-OH, 2’-NH2, or uracil, e.g., to a ribose.
32. The method of any of paragraphs 26-31, wherein the nucleotide modification is located
internally or at the 3’ end of the primer.
33. The method of any of paragraphs 23-32, wherein at least one primer comprises (i) a first
member; (ii) a second member; and ally (iii) a nucleotide modification described herein, e.g.,
located between (i) and (ii).
34. The method of paragraph 33, wherein the first member is capable of annealing with the
second member in the same primer or a different primer, e.g., forming a hairpin structure (via
intramolecular hybridization) or a double-stranded structure (via intermolecular hybridization)
sing a duplex region of 4, 5, 6, 7, 8, 9, 10, ll, 12, 13, 14, 15, 16, 17, 18, 19, 20, more
basepairs.
. The method of paragraph 33 or 34, wherein the first member comprises a ce that is
complementary to the sequence of an ucleotide attached to the capture substrate.
36. The method of any of aphs 33-35, wherein the second member comprises (e.g.,
from 5’ to 3’) one, two, or all of: (i) a sequence that is complementary to at least a portion of the first
member; (ii) a universal priming sequence (e.g., for PCR amplification or next-generation
sequencing); and (iii) a sequence complementary to a target sequence, e.g., an HCVRS and/or a
LCVRS, optionally, wherein the second member comprises a sequence for homologous
recombination (e.g., in a yeast or ian cell).
37. The method of any of paragraphs 23-36, wherein at least one primer comprises a
sequence encoding at least a portion of a linker ce, or a complementary sequence thereof.
38. The method of paragraph 37, wherein the primer that comprises a sequence encoding at
least a portion of a linker sequence, or a complementary sequence thereof, is phosphorylated, e.g., 5’
orylated.
39. The method of paragraph 37 or 38, wherein the linker sequence comprises, or consists of,
(Gly-Gly-Gly-Gly-Ser)n, where n=1, 2, 3, 4, 5, or more.
40. The method of any of paragraphs 1-39, wherein the HC ds cDNA comprises a 5’
overhang, e.g., a 5’ overhang that is capable of izing to an oligonucleotide attached to a capture
substrate.
41. The method of any of paragraphs 1-40, wherein the HC ds cDNA comprises a blunt end,
e.g., a blunt end comprising a 5’ phosphate.
42. The method of any of paragraphs 1-41, wherein the LC ds cDNA comprises a 5’
overhang, e.g., a 5’ overhang that is capable of hybridizing to an oligonucleotide attached to a capture
substrate.
43. The method of any of paragraphs 1-42, wherein the LC ds cDNA ses a blunt end,
e.g., a blunt end comprising a 5’ phosphate.
44. The method of any of paragraphs 1-43, n the HC ds cDNA and the LC ds cDNA
comprise sticky ends, e.g., both have 5’ ngs.
45. The method of any of paragraphs 1-44, wherein the HC strand and the LC strand are
covalently linked, e.g., ligated, to produce a single stranded nucleic acid sequence, n the HC
and LC strands are both sense strands or both antisense strands.
46. The method of any of paragraphs 1-44, n a denatured HC strand of the HC ds
cDNA to a denatured LC strand of the LC ds cDNA are covalently , e.g., ligated, wherein the
HC and LC strands are both sense strands or both antisense strands.
47. The method of any of paragraphs 1-44, wherein the HC strand is present in the HC ds
cDNA and the LC strand is present in the LC ds cDNA, and wherein the HC ds cDNA and the LC ds
cDNA are covalently linked, e.g., ligated, e.g., to produce a double stranded nucleic acid sequence.
48. The method of any of paragraphs 1-47, wherein the covalent linking, e.g., ligation, occurs
in the isolated production reaction site.
49. The method of paragraph 48, wherein the isolated production reaction site, e.g., a
production micro-chamber, comprises a reagent that is capable of covalently linking, e.g., ligating, the
HC and LC strands or the HC and LC ds cDNAs.
50. The method of paragraph 48 or 49, wherein the isolated production reaction site, e.g., a
production micro-chamber comprises an enzyme that covalently couples the HC and LC strands or the
HC and LC ds cDNAs.
51. The method of any of paragraphs 1-47, wherein the covalent linking, e.g., ligation, occurs
in a site different from the isolated production reaction site, e.g., occurs in an isolated e reaction
site, e.g., a linkage micro-chamber.
52. The method of paragraph 51, wherein the HC strand and the LC strand are erred
from the isolated production site to the isolated e reaction site, e.g., a e micro-chamber,
and the covalent linking occurs in the isolated linkage reaction site, e.g., a linkage micro-chamber.
53. The method of paragraph 51 or 52, wherein the ed linkage reaction site, e.g., a
linkage micro-chamber, comprises a reagent that is capable of covalently linking, e.g., ligating, the
HC and LC strands or the HC and LC ds cDNAs.
54. The method of any of paragraphs 51-53, wherein the isolated linkage reaction site, e.g., a
linkage chamber, comprises an enzyme that covalently couples the HC and LC strands or the
HC and LC ds cDNAs.
55. The method of paragraph 50 or 54, wherein the enzyme is a ligase, e.g., a thermal stable
56. The method of any of paragraphs 51-55, wherein the covalent linking, e.g., ligation,
comprises:
(a) heating the isolated linkage on site, e.g., the linkage micro-chamber, under
ions (e.g., at 95°C) that allow denaturation of the HC strand and the LC strand;
(b) g the isolated linkage reaction site, e.g., the linkage micro-chamber, under
conditions (e.g., at 50-65°C) that allow hybridization of the splint oligonucleotide to the HC strand
and the LC strand;
(c) maintaining the isolated linkage reaction site, e.g., the linkage micro-chamber, under
conditions (e.g., at 45-65°C) that allow on of the HC strand and the LC strand (e.g., formation of
phosphodiester bond between the HC strand and the LC strand); and
(d) repeating steps (a), (b), and (c) sequentially for 2, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, or
more cycles.
57. The method of any of paragraphs 1-56, wherein the HC strand and the LC strand are
covalently linked, e.g., d, in the presence of a splint oligonucleotide.
58. The method of paragraph 57, wherein the splint oligonucleotide is hybridized to a
sequence comprising the junction of the HC strand and the DC strand, or a sequence complementary
thereof, and forms a duplexed region at the site of ligation.
59. The method of paragraph 57 or 58, wherein the splint oligonucleotide comprises a
modification (e.g., an NH2 group) that inhibits DNA synthesis, e.g., by a DNA polymerase.
60. The method of paragraph 59, wherein the cation is at the 3’ end of the splint
oligonucleotide.
61. The method of any of paragraphs 1-60, wherein a strand complimentary to the covalently
, e.g., ligated, HC and LC strands is produced by amplification.
62. The method of any of paragraphs 1-61, further comprising, prior to acquiring the isolated
production reaction site, e.g., a production micro-chamber, acquiring an mRNA loaded capture
substrate.
63. The method of paragraph 62, wherein acquiring the mRNA loaded capture substrate
sing:
a) acquiring an isolated cell reaction site, e.g., a cell isolation chamber, comprising:
i) a cell; and
ii) a e substrate capable of binding a first mRNA encoding an HCVR from the
cell and a second mRNA encoding an LCVR from the cell; and
b) maintaining the isolated cell reaction site, e.g., the cell isolation micro-chamber, under
conditions that allow lysis of the cell and binding of the capture substrate with the first mRNA and the
second mRNA to form the mRNA loaded capture substrate,
wherein the isolated cell reaction site, e.g., cell isolation micro-chamber, does not include a
nucleic acid encoding an HCVR or an LCVR from a cell other than the cell.
64. The method of aph 63, wherein the isolated cell reaction site, e. g., cell isolation
chamber, ses a lysing reagent, e.g., a detergent.
65. The method of paragraph 63 or 64, n the cell is lysed by heat or an enzyme.
66. The method of any of paragraphs 63-65, wherein the capture ate comprises a
moiety (e.g., an oligonucleotide) which binds mRNA, e.g., an oligo(dT).
67. The method of any of paragraphs 62-66, further comprising releasing the mRNA loaded
capture substrate from the isolated cell reaction site, e.g., the cell isolation micro-chamber.
68. The method of paragraph 67, wherein the releasing step is performed in the presence of a
poly(dA) or T) oligonucleotide, e.g., to reduce cross-binding of non-captured mRNA.
69. The method of paragraph 62-68, wherein the mRNA loaded capture substrate is
transferred from the isolated cell reaction site, e.g., the cell isolation micro-chamber, to the isolated
production reaction site, e.g., the production micro-chamber.
70. The method of any of paragraphs 1-69, comprising releasing the nucleic acid sequence
from the production micro-chamber.
71. The method of any of paragraphs 1-70, further comprising amplifying the nucleic acid
sequence.
72. The method of any of paragraphs 1-71, comprising cing all or a portion of the
nucleic acid sequence.
73. The method of any of paragraphs 1-72, sing inserting all or a portion of c
acid sequence into a .
74. The method of paragraph 73, wherein the vector supplies an onal HC element or LC
element not included in the nucleic acid sequence.
75. The method of paragraph 73 or 74, wherein the vector supplies an HC CDRl, an HC
CDRZ, or both.
76. The method of any of paragraphs 73-75, comprising sing the vector.
77. The method of any of aphs 1-76, comprising expressing the nucleic acid sequence
to produce a polypeptide comprising a segment that encodes an HC element of the HCVR, e.g., an
HCVRS, and a segment that encodes an LC element of the LCVR, e.g., an LCVRS.
78. The method of paragraph 77, wherein the LC t is inal to the HC element in
the polypeptide.
79. The method of paragraph 77, wherein the HC element is C-terminal to the LC t in
the polypeptide.
80. The method of any of paragraphs 77-79, further comprising contacting the polypeptide
with an antigen.
81. The method of any of paragraphs 77-80, further comprising determining if the
polypeptide binds the antigen.
82. The method of any of paragraphs 1-81, wherein the cell is an immune cell, e.g., a B cell
or T cell, e. g., a human B cell or T cell.
83. The method of any of paragraphs 1-82, wherein the cell is a mammalian cell or an avian
cell.
84. A method of making a nucleic acid sequence comprising a ce that encodes a heavy
chain element (HC element) of an antibody heavy chain variable region (HCVR) and a light chain
element (LC element) of an antibody light chain variable region (LCVR), and wherein the HCVR and
LCVR are matched, comprising:
a) acquiring an isolated cell reaction site, e.g., a cell isolation micro-chamber, comprising:
i) a cell; and
ii) a capture substrate capable of binding a first mRNA encoding an HCVR from the
cell and a second mRNA encoding an LCVR from the cell;
b) maintaining isolated cell reaction site, e.g., the cell isolation micro-chamber, under
conditions that allow lysis of the cell and binding of the capture substrate with the first mRNA and the
second mRNA to form the mRNA loaded capture ate,
wherein the isolated cell reaction site, e.g., cell ion micro-chamber, does not e a
nucleic acid encoding an HCVR or an LCVR from a cell other than the cell;
c) contacting the mRNA loaded capture substrate with a on mixture, e.g., a reaction
mixture comprising reverse transcriptase, that uses the loaded mRNA as a te to make cDNA
(this can occur, e.g., in the isolated cell reaction site, in the isolated production reaction site, or in
neither, e.g., not in an isolated reaction site);
d) acquiring an isolated production reaction site, e.g., a production micro-chamber,
comprising:
i) a heavy chain (HC) strand from step b), wherein the HC strand is a strand of a
heavy chain double-stranded cDNA (HC ds cDNA) comprising a segment that s an HC
element of the HCVR from the cell, e.g., a heavy chain le region sequence );
ii) a light chain (LC) strand from step b), wherein the LC strand is a strand of a light
chain double-stranded cDNA (LC ds cDNA) comprising a segment that encodes an LC
element of the LCVR from the cell, e.g., a light chain variable region sequence (LCVRS); and
e) covalent linking, e.g., ligation, of an HC strand to an LC strand,
wherein the isolated production reaction site, e.g., a production micro-chamber, does not
include a nucleic acid encoding an LCVR or an HCVR from a cell other than the cell.
85. A method of making a nucleic acid sequence comprising a sequence that encodes a heavy
chain element (HC element) of an antibody heavy chain variable region (HCVR) and a light chain
element (LC element) of an antibody light chain variable region (LCVR), and wherein the HCVR and
LCVR are matched, comprising:
a) acquiring an isolated cell reaction site, e.g., a cell ion chamber, comprising:
i) a cell; and
ii) a e substrate capable of binding a first mRNA encoding an HCVR from the
cell and a second mRNA encoding an LCVR from the cell;
b) maintaining the isolated cell reaction site, e.g., the cell isolation micro-chamber, under
conditions that allow lysis of the cell and binding of the capture substrate with the first mRNA and the
second mRNA to form the mRNA loaded capture substrate,
wherein the isolated cell reaction site, e.g., cell isolation chamber, does not include a
nucleic acid ng an HCVR or an LCVR from a cell other than the cell;
c) acquiring an isolated production reaction site, e.g., a production micro-chamber, comprises:
contacting the mRNA loaded capture substrate with a reaction mixture, e.g., a reaction mixture
comprising reverse transcriptase, that uses the loaded mRNA as a template, to produce:
a first double-stranded cDNA (ds cDNA) comprising a strand that is complementary to a first
mRNA that s an HCVR from a cell; and
a second ds cDNA comprising a strand complementary to a second mRNA encoding an
LCVR from the cell (the cDNA loaded capture substrate);
wherein the ed production reaction site, e.g., a production micro-chamber, does not
include a nucleic acid encoding an LCVR or an HCVR from a cell other than the cell.
d) ining the isolated production reaction site, e.g., the production micro-chamber,
under conditions that allow amplification of the first and second ds cDNAs, to produce:
a plurality of HC ds cDNAs comprising a segment that encodes an HC element of the HCVR
from the cell, e.g., an HCVRS; and
a plurality of LC ds cDNAs sing a segment that encodes an LC element of the LCVR
from the cell, e. g., an LCVRS;
e) acquiring an isolated linkage reaction site, e.g., a e micro-chamber, comprising:
covalent linking, e.g., ligation, of a strand of the HC ds cDNA (HC strand) to a strand of the LC ds
cDNA (LC strand), wherein the HC and LC strands are both sense strands or antisense strands; and
f) amplifying the covalently linked, e.g., ligated, HC and LC strands.
86. A method of making a y sing a plurality of unique members, the method
comprising:
making the plurality of members, wherein each of the s comprises a sequence that
s a heavy chain element (HC t) of a heavy chain variable region (HCVR) and a light
chain element (LC element) of a light chain le region (LCVR), and wherein the HCVR and
LCVR are matched, made by a method of any of paragraphs 1-85,
wherein each unique nucleic acid ce of the plurality comprises an HC element and an
LC element from a different unique cell,
thereby making a library comprising a plurality of unique members.
87. The method of paragraph 86, wherein the plurality of unique members comprises at least
104, 105, 106, 107, 108, or 109 unique members.
2017/068204
88. The method of paragraph 86 or 87, wherein the plurality of unique members comprises
104 to 109, 104 to 108, 104 to 107, 104 to 106, 104 to 105, 108 to 109, 107 to 109, 106 to 109, 105 to 109,
105 to 108, 106 to 107, 104 to 105, 105 to 106, 106 to 107, 107 to 108, or 108 to 109 unique members.
89. The method of any of paragraphs 86-88, wherein at least 80%, 85 %, 90%, 95%, 96%,
97%, 98%, 99%, or 100%, of the s in the library are unique members (which encode matched
HC element and LC element sequences).
90. The method of any of paragraphs 86-89, wherein less than 20%, 15%, 10%, 5%, 4%, 3%,
2%, or 1%, of the members in the library are unique members (which encode matched HC element
and LC element sequences).
91. A library comprising:
a plurality of unique members,
wherein,
i) each unique member of the plurality comprises a segment that s an HC element, e.g.,
an HCVRS, and a segment that encodes an LC element, e.g., an LCVRS, wherein the HC element and
the LC element in each unique member is matched;
ii) each unique member of the plurality comprises a segment that encodes an HC element,
e.g., an HCVRS, and a segment that encodes an LC element, e.g., an LCVRS, from a different unique
cell; and
iii) the y comprises one or more (e.g., two, three, four, or all) of the following
properties:
a) the library is made by a method of any of paragraphs 1-85;
b) the plurality of unique members comprises at least 104, 105, 106, 107, 108, or 109
unique nucleic acid sequences;
c) the ity of unique members comprises 104 to 109, 104 to 108, 104 to 107, 104 to
106, 104 to 105, 108 to 109, 107 to 109, 106 to 109, 105 to 109, 105 to 108, 106 to 107, 104 to 105,
105 to 106, 106 to 107, 107 to 108, or 108 to 109 unique members;
(1) at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%, of the members in
the library are unique members (which encode matched HC element and LC t
sequences); or
e) less than 20%, 15%, 10%, 5%, 4%, 3%, 2%, or 1%, of the members in the library
are unique members (which encode matched HC t and LC element sequences).
92. The library of paragraph 91, wherein each unique member of the plurality is ured
such that, when expressed, the HC element, e.g., the HCVRS, and the LC element, e. g., the LCVRS,
form a functional antigen binding molecule, e.g., an scFV.
93. The library of any of paragraphs 91-92, n the library is a display library.
94. The library of any of paragraphs 91-93, wherein each of the members of the ity
further s a polypeptide that results in display of the member on the surface of a display entity.
95. The library of any of paragraphs 91-94, wherein the library is a phage display library.
96. The library of any of paragraphs 91-94, n the library is a yeast display library.
97. The library of any of paragraphs 91-94, n the library is a mammalian display
library.
98. A method of making a binding polypeptide, the method sing:
a) acquiring a library of any of paragraphs 91-97; and
b) expressing a polypeptide encoded by a unique c acid of the library.
99. The method of paragraph 98, further comprising contacting the polypeptide with an
antigen.
100. The method of paragraph 98 or 99, further comprising ving the nucleic acid that
encodes a polypeptide that binds the antigen.
101. A method of making a nucleic acid sequence comprising a sequence that encodes an 0:
chain element (AC element) of a TCR 0t chain variable region (ACVR) and a [3 chain element (BC
element) of a TCR [3 chain variable region (BCVR), and wherein the ACVR and BCVR are matched,
the method comprising:
a) acquiring an isolated production reaction site, e.g., a production micro-chamber,
comprising:
i) an a chain (AC) strand, wherein the AC strand is a strand of an 0. chain double-
ed cDNA (AC ds cDNA) comprising a t that encodes an AC element of the
ACVR from a cell, e.g., an a chain variable region ce (ACVRS); and
ii) a [5 chain (BC) strand, wherein the BC strand is a strand of a B chain double-
ed cDNA (BC ds cDNA) comprising a segment that encodes a BC t of the
BCVR from the cell, e.g., a [3 chain variable region sequence (BCVRS), and
b) covalent linking, e.g., ligation, of an AC strand to a BC strand,
n the isolated production reaction site, e.g., a production micro-chamber, does not
include a nucleic acid encoding a BCVR or an ACVR from a cell other than the cell,
thereby making a nucleic acid sequence comprising a sequence that encodes an AC element
of an ACVR and a BC element of a BCVR, wherein the ACVR and BCVR are matched.
102. The method of paragraph 101, wherein the AC element comprises, or consists of, an
ACVRS, or a functional fragment f (e.g., an antigen binding fragment thereof).
103. The method of paragraph 101 or 102, wherein the BC element comprises, or ts of,
a BCVRS, or a functional fragment thereof (e.g., an antigen g fragment thereof).
104. The method of any of paragraphs 101-103, wherein the nucleic acid sequence is
configured such that, when expressed, the ACVRS and the BCVRS form a functional antigen binding
le, e. g., a single chain TCR molecule.
105. The method of paragraph 104, wherein the antigen binding molecule is functional in
vitro, ex vivo, or in viva, e.g., as determined by a method or assay described herein.
106. The method of any of paragraphs 101-105, wherein acquiring an isolated production
reaction site, e.g., a production micro-chamber, comprises:
a) acquiring a capture ate bound to:
(i) a first double-stranded cDNA (ds cDNA) sing a strand that is
complementary to a first mRNA that encodes an ACVR from a cell; and
(ii) a second ds cDNA comprising a strand complementary to a second mRNA
encoding a BCVR from the cell (the cDNA loaded capture substrate), and
b) maintaining the isolated production reaction site, e.g., the production micro-chamber,
under conditions that allow amplification of the first and second ds cDNAs, to e:
a ity of AC ds cDNAs comprising a segment that encodes an AC element of the ACVR
from the cell, e.g., an ACVRS; and
a plurality of BC ds cDNAs comprising a segment that encodes a BC element of the BCVR
from the cell, e.g., a BCVRS.
107. The method of paragraph 106, wherein the AC ds cDNA is identical, or substantially
identical, to the first ds cDNA.
108. The method of aph 106 or 107, wherein the BC ds cDNA is identical, or
substantially identical, to the second ds cDNA.
109. The method of any of paragraphs 106-108, wherein the capture substrate ses a
bead, e.g., a magnetic bead.
110. The method of any of paragraphs 106-109, wherein the capture ate comprises a
moiety (e.g., an oligonucleotide) which binds to cDNA, e.g., (i) a moiety which binds to the AC
strand; (ii) a moiety which binds to the BC strand; or (iii) both (i) and (ii).
111. The method of any of paragraphs 106-110, wherein the first mRNA and the second
mRNA are disposed on an mRNA loaded capture ate.
112. The method of any of paragraphs 1, wherein the isolated production reaction site,
e.g., the production micro-chamber, comprises:
a reagent mixture suitable for producing, from the first and second mRNAs (e.g., after the
first and second mRNAs are released from the mRNA loaded capture substrate into a solution), a first
ds cDNA comprising a segment that encodes an AC element of the ACVR of the cell, e. g., an
ACVRS, and a second ds cDNA comprising a segment that encodes a BC element of the BCVR of the
cell, e.g., a BCVRS.
113. The method of any of aphs 2, wherein the isolated production reaction site,
e.g., production micro-chamber, comprises primers that mediate the production of the first ds cDNA.
114. The method of any of paragraphs 106-113, wherein the isolated production reaction site,
e.g., production micro-chamber, comprises s that mediate the production of the second ds
cDNA.
115. The method of any of paragraphs 106-114, wherein a cDNA strand that is
complementary to a first mRNA that encodes an ACVR from a cell is made by e transcription
of the first mRNA.
116. The method of any of aphs 106-115, n a cDNA strand that is
complementary to a second mRNA that encodes a BCVR from a cell is made by reverse transcription
of the second mRNA.
117. The method of paragraph 115 or 116, wherein the reverse transcription takes place in
the isolated production reaction site, e.g., a production-micro chamber.
118. The method of paragraph 115 or 116, n the reverse transcription takes place in an
isolated cell reaction site, e.g., a cell isolation micro-chamber.
119. The method of paragraph 115 or 116, wherein the reverse transcription takes place
outside the isolated production reaction site, e.g., a production chamber, or outside an isolated
cell reaction site, e.g., a cell isolation chamber.
120. The method of paragraph 115 or 116, wherein the reverse transcription takes place
outside the isolated production reaction site, e.g., a production-micro chamber, and outside an isolated
cell reaction site, e.g., a cell isolation chamber.
121. The method of paragraph 115 or 116, wherein the reverse transcription takes place
outside an isolated reaction site, e.g., outside a micro-chamber.
122. The method of any of paragraphs 106-121, wherein the amplification comprises 20 or
fewer cycles, e.g., 15 or fewer, 14 or fewer, 13 or fewer, 12 or fewer, 11 or fewer, 10 or fewer, 9 or
fewer, 8 or fewer, 7 or fewer, 6 or fewer, or 5 or fewer cycles.
123. The method of any of aphs 106-122, wherein the reverse transcription and/or
amplification uses one or more primers, e.g., comprising a sequence specific for an ACVRS and/or a
BCVRS.
124. The method of any of aphs 106-123, n the amplification comprises using
two or more primers that mediate the production of the AC ds cDNA, wherein at least one primer
ses a nucleotide modification, and wherein at least one primer does not comprise a nucleotide
modification.
125. The method of any of paragraphs 106-124, wherein the amplification ses using
two or more primers that mediate the tion of the BC ds cDNA, wherein at least one primer
comprises a nucleotide modification, and wherein at least one primer does not comprise a nucleotide
modification.
126. The method of paragraph 125, wherein at least one primer comprises a nucleotide
modification, e.g., which reduces, e.g., inhibits, DNA synthesis, e.g., by a DNA polymerase.
127. The method of paragraph 125 or 126, wherein at least one primer does not comprise a
nucleotide modification, e.g., which reduces, e.g., inhibits, DNA synthesis, e.g., by a DNA
polymerase.
128. The method of paragraph 126 or 127, wherein the nucleotide modification inhibits a
DNA polymerase from extending the DNA.
129. The method of any of paragraphs 126-128, wherein the nucleotide modification is an
insertion of a spacer to the primer, e.g., between two adjacent tides in the primer.
130. The method of paragraph 129, wherein the spacer is a flexible spacer, a carbon spacer
(e.g., -(CH2)n-, wherein n=3, 4, 5, or more), two or more (e.g., three, four, five, or more) abasic
nucleotides or a PEG .
131. The method of any of paragraphs 126-128, wherein the nucleotide modification is 2’-O-
methyl, 2’-OH, 2’-NH2, or , e.g., to a ribose.
132. The method of any of paragraphs 126-131, wherein the nucleotide modification is
d internally or at the 3’ end of the primer.
133. The method of any of paragraphs 123-132, wherein at least one primer comprises (i) a
first member; (ii) a second ; and optionally (iii) a nucleotide modification described herein,
e. g., located between (i) and (ii).
134. The method of paragraph 133, n the first member is capable of ing with the
second member in the same primer or a different primer, e.g., forming a hairpin structure (via
intramolecular hybridization) or a double-stranded structure (via intermolecular hybridization)
comprising a duplex region of 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, more
basepairs.
135. The method of paragraph 133 or 134, wherein the first member comprises a ce
that is complementary to the ce of an oligonucleotide attached to the capture substrate.
136. The method of any of paragraphs 133-135, wherein the second member comprises (e.g.,
from 5’ to 3’) one, two, or all of: (i) a sequence that is complementary to at least a portion of the first
member; (ii) a universal priming sequence (e.g., for PCR amplification or next-generation
sequencing); and (iii) a sequence complementary to a target sequence, e. g., an ACVRS and/or a
BCVRS, optionally, wherein the second member comprises a sequence for homologous
recombination (e.g., in a yeast or mammalian cell).
137. The method of any of paragraphs 6, wherein at least one primer ses a
sequence encoding at least a portion of a linker sequence, or a complementary sequence thereof.
138. The method of paragraph 137, wherein the primer that comprises a sequence encoding
at least a n of a linker ce, or a complementary sequence thereof, is phosphorylated, e.g.,
’ phosphorylated.
139. The method of paragraph 137 or 138, wherein the linker sequence comprises, or consists
of, (Gly-Gly-Gly-Gly-Ser)n, where n=1, 2, 3, 4, 5, or more.
140. The method of any of paragraphs 101-139, wherein the AC ds cDNA comprises a 5’
overhang, e.g., a 5’ overhang that is capable of hybridizing to an oligonucleotide attached to a capture
substrate.
141. The method of any of paragraphs 101-140, wherein the AC ds cDNA comprises a blunt
end, e.g., a blunt end comprising a 5’ phosphate.
142. The method of any of paragraphs 101-141, wherein the BC ds cDNA ses a 5’
overhang, e.g., a 5’ ng that is capable of hybridizing to an oligonucleotide attached to a capture
substrate.
143. The method of any of paragraphs 2, wherein the BC ds cDNA comprises a blunt
end, e.g., a blunt end comprising a 5’ phosphate.
144. The method of any of paragraphs 101-143, n the AC ds cDNA and the BC ds
cDNA comprise sticky ends, e.g., both have 5’ overhangs.
145. The method of any of paragraphs 101-144, wherein the AC strand and the BC strand are
covalently linked, e.g., ligated, to e a single stranded nucleic acid sequence, wherein the AC
and BC strands are both sense strands or both antisense strands.
146. The method of any of paragraphs 101-144, wherein a denatured AC strand of the AC ds
cDNA to a red BC strand of the BC ds cDNA are covalently linked, e.g., ligated, wherein the
AC and BC strands are both sense strands or both antisense strands.
147. The method of any of paragraphs 101-144, wherein the AC strand is t in the AC
ds cDNA and the BC strand is t in the BC ds cDNA, and wherein the AC ds cDNA and the BC
ds cDNA are covalently linked, e.g., ligated, e. g., to produce a double stranded nucleic acid sequence.
148. The method of any of paragraphs 101-147, n the covalent g, e.g., ligation,
occurs in the isolated production reaction site.
149. The method of paragraph 148, wherein the isolated production reaction site, e.g., a
production micro-chamber, comprises a reagent that is capable of covalently linking, e. g., ligating, the
AC and BC strands or the AC and BC ds cDNAs.
150. The method of paragraph 148 or 149, wherein the isolated production reaction site, e.g.,
a production micro-chamber comprises an enzyme that covalently couples the AC and BC strands or
the AC and BC ds cDNAs.
151. The method of any of paragraphs 101-147, wherein the covalent linking, e. g., ligation,
occurs in a site different from the isolated production reaction site, e.g., occurs in an isolated linkage
reaction site, e.g., a e micro-chamber.
152. The method of paragraph 151, wherein the AC strand and the BC strand are transferred
from the isolated production site to the isolated linkage reaction site, e.g., a linkage micro-chamber,
and the covalent linking occurs in the isolated linkage reaction site, e.g., a e micro-chamber.
153. The method of paragraph 151 or 152, wherein the isolated linkage reaction site, e.g., a
linkage micro-chamber, ses a t that is capable of covalently linking, e.g., ligating, the
AC and BC strands or the AC and BC ds cDNAs.
WO 19402
154. The method of any of paragraphs 151-153, wherein the isolated linkage reaction site,
e.g., a linkage micro-chamber, comprises an enzyme that covalently couples the AC and BC strands
or the AC and BC ds cDNAs.
155. The method of paragraph 150 or 154, wherein the enzyme is a ligase, e.g., a thermal
stable ligase.
156. The method of any of paragraphs 151-155, n the covalent linking, e.g., ligation,
ses:
(a) heating the isolated linkage reaction site, e.g., the linkage micro-chamber, under
conditions (e.g., at 95°C) that allow ration of the AC strand and the BC strand;
(b) g the isolated linkage reaction site, e.g., the linkage micro-chamber, under
conditions (e.g., at 50-65°C) that allow hybridization of the splint oligonucleotide to the AC strand
and the BC strand;
(c) maintaining the isolated linkage reaction site, e.g., the e micro-chamber, under
conditions (e.g., at 45-65°C) that allow ligation of the AC strand and the BC strand (e.g., formation of
phosphodiester bond between the AC strand and the BC strand); and
(d) repeating steps (a), (b), and (c) sequentially for 2, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, or
more cycles.
157. The method of any of paragraphs 101-156, n the AC strand and the BC strand are
covalently linked, e.g., ligated, in the presence of a splint oligonucleotide.
158. The method of paragraph 157, wherein the splint ucleotide is hybridized to a
sequence comprising the junction of the AC strand and the BC strand, or a sequence complementary
thereof, and forms a duplexed region at the site of ligation.
159. The method of paragraph 157 or 158, wherein the splint oligonucleotide comprises a
modification (e.g., an NH2 group) that inhibits DNA synthesis, e.g., by a DNA polymerase.
160. The method of paragraph 159, wherein the ation is at the 3’ end of the splint
oligonucleotide.
161. The method of any of paragraphs 101-160, n a strand complimentary to the
covalently linked, e.g., ligated, AC and BC strands is produced by amplification.
162. The method of any of paragraphs 101-161, further comprising, prior to acquiring the
isolated production reaction site, e.g., a production micro-chamber, acquiring an mRNA loaded
capture substrate.
163. The method of paragraph 162, wherein acquiring the mRNA loaded capture substrate
comprising:
a) acquiring an isolated cell reaction site, e.g., a cell ion micro-chamber, sing:
i) a cell; and
ii) a capture substrate capable of binding a first mRNA encoding an ACVR from the
cell and a second mRNA encoding a BCVR from the cell; and
b) maintaining the isolated cell reaction site, e.g., the cell isolation micro-chamber, under
conditions that allow lysis of the cell and binding of the e substrate with the first mRNA and the
second mRNA to form the mRNA loaded capture substrate,
wherein the ed cell on site, e.g., cell isolation chamber, does not include a
nucleic acid encoding an ACVR or a BCVR from a cell other than the cell.
164. The method of paragraph 163, wherein the isolated cell reaction site, e.g., cell isolation
micro-chamber, comprises a lysing reagent, e.g., a detergent.
165. The method of aph 163 or 164, wherein the cell is lysed by heat or an enzyme.
166. The method of any of paragraphs 163-165, wherein the capture substrate comprises a
moiety (e.g., an oligonucleotide) which binds mRNA, e.g., an oligo(dT).
167. The method of any of paragraphs 162-166, further comprising releasing the mRNA
loaded capture substrate from the isolated cell reaction site, e.g., the cell isolation chamber.
168. The method of paragraph 167, wherein the releasing step is performed in the presence of
a poly(dA) or poly(dT) oligonucleotide, e.g., to reduce cross-binding of non-captured mRNA.
169. The method of paragraph 162-168, wherein the mRNA loaded capture substrate is
transferred from the isolated cell reaction site, e.g., the cell isolation micro-chamber, to the isolated
tion reaction site, e.g., the production micro-chamber.
170. The method of any of aphs 101-169, comprising ing the c acid
sequence from the tion chamber.
171. The method of any of paragraphs 101-170, further comprising amplifying the nucleic
acid sequence.
172. The method of any of paragraphs 101-171, comprising sequencing all or a portion of the
nucleic acid sequence.
173. The method of any of paragraphs 101-72, comprising inserting all or a portion of
nucleic acid sequence into a .
174. The method of paragraph 173, wherein the vector supplies an additional AC element or
BC t not included in the nucleic acid sequence.
175. The method of paragraph 173 or 174, wherein the vector supplies an AC CDRl, an AC
CDR2, or both.
176. The method of any of paragraphs 173-175, comprising sing the vector.
177. The method of any of paragraphs 101-176, comprising expressing the nucleic acid
sequence to produce a polypeptide comprising a segment that encodes an AC element of the ACVR,
e. g., an ACVRS, and a segment that encodes a BC element of the BCVR, e. g., a BCVRS.
178. The method of paragraph 177, wherein the BC element is N—terminal to the AC element
in the polypeptide.
179. The method of paragraph 177, wherein the AC element is C-terminal to the BC element
in the polypeptide.
180. The method of any of paragraphs 177-179, further comprising contacting the
polypeptide with an antigen.
181. The method of any of paragraphs 0, further comprising determining if the
polypeptide binds the antigen.
182. The method of any of paragraphs 101-181, wherein the cell is an immune cell, e.g., a B
cell or T cell, e.g., a human B cell or T cell.
183. The method of any of paragraphs 101-182, wherein the cell is a mammalian cell or an
avian cell.
184. A method of making a nucleic acid sequence comprising a sequence that encodes an 0:
chain t (AC element) of a TCR 0t chain variable region (ACVR) and a [3 chain element (BC
element) of a TCR [3 chain variable region (BCVR), and n the ACVR and BCVR are matched,
comprising:
a) acquiring an isolated cell reaction site, e.g., a cell isolation micro-chamber, sing:
i) a cell; and
ii) a capture substrate capable of binding a first mRNA encoding an ACVR from the
cell and a second mRNA encoding a BCVR from the cell;
b) ining isolated cell reaction site, e.g., the cell isolation micro-chamber, under
conditions that allow lysis of the cell and binding of the capture substrate with the first mRNA and the
second mRNA to form the mRNA loaded capture substrate,
wherein the isolated cell on site, e. g., cell isolation micro-chamber, does not include a
nucleic acid encoding an ACVR or a BCVR from a cell other than the cell;
c) contacting the mRNA loaded capture substrate with a reaction mixture, e.g., a reaction
mixture comprising reverse riptase, that uses the loaded mRNA as a template to make cDNA
(this can occur, e.g., in the isolated cell reaction site, in the ed tion reaction site, or in
neither, e.g., not in an ed reaction site);
d) acquiring an isolated production reaction site, e.g., a production micro-chamber,
comprising:
i) an on chain (AC) strand from step b), wherein the AC strand is a strand of an 0t chain
double-stranded cDNA (AC ds cDNA) comprising a segment that s an AC element of
the ACVR from the cell, e.g., a 0L chain variable region sequence (ACVRS); and
ii) a [3 chain (BC) strand from step b), wherein the BC strand is a strand of a [3 chain
double-stranded cDNA (BC ds cDNA) comprising a segment that encodes a BC element of
the BCVR from the cell, e.g., a B chain variable region sequence (BCVRS); and
e) covalent linking, e.g., ligation, of an AC strand to a BC strand,
wherein the isolated production reaction site, e.g., a production micro-chamber, does not
include a nucleic acid encoding a BCVR or an ACVR from a cell other than the cell.
185. A method of making a nucleic acid sequence sing a sequence that encodes a or
chain element (AC element) of an TCR or chain variable region (ACVR) and a [3 chain element (BC
element) of an TCR B chain le region (BCVR), and wherein the ACVR and BCVR are matched,
comprising:
a) acquiring an isolated cell reaction site, e.g., a cell isolation micro-chamber, comprising:
i) a cell; and
ii) a capture substrate capable of binding a first mRNA encoding an ACVR from the
cell and a second mRNA encoding a BCVR from the cell;
b) maintaining the isolated cell reaction site, e.g., the cell isolation micro-chamber, under
conditions that allow lysis of the cell and binding of the capture substrate with the first mRNA and the
second mRNA to form the mRNA loaded capture substrate,
n the isolated cell reaction site, e.g., cell isolation micro-chamber, does not include a
c acid encoding an ACVR or a BCVR from a cell other than the cell;
c) acquiring an isolated production reaction site, e. g., a production micro-chamber, ses:
contacting the mRNA loaded capture substrate with a reaction mixture, e.g., a reaction mixture
comprising reverse transcriptase, that uses the loaded mRNA as a template, to produce:
a first double-stranded cDNA (ds cDNA) comprising a strand that is complementary to a first
mRNA that encodes an ACVR from a cell; and
a second ds cDNA comprising a strand complementary to a second mRNA encoding a BCVR
from the cell (the cDNA loaded capture ate);
wherein the ed production reaction site, e.g., a production micro-chamber, does not
e a nucleic acid encoding a BCVR or an ACVR from a cell other than the cell.
d) maintaining the isolated production reaction site, e.g., the production micro-chamber,
under conditions that allow amplification of the first and second ds cDNAs, to produce:
a plurality of AC ds cDNAs comprising a t that encodes an AC element of the ACVR
from the cell, e.g., an ACVRS; and
a plurality of BC ds cDNAs comprising a segment that encodes a BC element of the BCVR
from the cell, e.g., a BCVRS;
e) acquiring an isolated linkage reaction site, e.g., a linkage micro-chamber, comprising:
covalent linking, e.g., ligation, of a strand of the AC ds cDNA (AC strand) to a strand of the BC ds
cDNA (BC strand), wherein the AC and BC s are both sense strands or antisense strands; and
f) amplifying the ntly linked, e.g., ligated, AC and BC strands.
186. A method of making a library sing a ity of unique members, the method
comprising:
making the plurality of members, n each of the members comprises a sequence that
encodes a 0t chain element (AC element) of a 0t chain variable region (ACVR) and a [3 chain element
(BC t) of a [3 chain variable region (BCVR), and wherein the ACVR and BCVR are matched,
made by a method of any of paragraphs 101-185,
wherein each unique nucleic acid sequence of the ity comprises an AC element and a
BC element from a different unique cell,
thereby making a library comprising a plurality of unique members.
187. The method of paragraph 186, wherein the plurality of unique members comprises at
least 104, 105, 106, 107, 108, or 109 unique members.
188. The method of paragraph 186 or 187, wherein the plurality of unique members
comprises 104 to 109, 104 to 108, 104 to 107, 104 to 106, 104 to 105, 108 to 109, 107 to 109, 106 to 109, 105
to 109, 105 to 108, 106 to 107, 104 to 105, 105 to 106, 106 to 107, 107 to 108, or 108 to 109 unique
members.
189. The method of any of paragraphs 186-188, wherein at least 80%, 85%, 90%, 95%, 96%,
97%, 98%, 99%, or 100%, of the members in the library are unique members (which encode matched
AC element and BC elements ces).
190. The method of any of paragraphs 9, wherein less than 20%, 15%, 10%, 5%, 4%,
3%, 2%, or 1%, of the members in the library are unique members (which encode matched AC
element and BC ts sequences).
191. A y comprising:
a plurality of unique members,
wherein,
i) each unique member of the plurality comprises a segment that encodes an AC element, e.g.,
an ACVRS, and a segment that encodes a BC element, e.g., a BCVRS, wherein the AC element and
the BC element in each unique member is matched;
ii) each unique member of the plurality comprises a segment that encodes an AC element,
e.g., an ACVRS, and a t that encodes a BC element, e.g., a BCVRS, from a different unique
cell; and
iii) the library comprises one or more (e.g., two, three, four, or all) of the following
properties:
a) the library is made by a method of any of aphs 101-185;
b) the plurality of unique members comprises at least 104, 105, 106, 107, 108, or 109
unique nucleic acid sequences;
c) the plurality of unique s comprises 104 to 109, 104 to 108, 104 to 107, 104 to
106, 104 to 105, 108 to 109, 107 to 109, 106 to 109, 105 to 109, 105 to 108, 106 to 107, 104 to 105,
105 to 106, 106 to 107, 107 to 108, or 108 to 109 unique members;
d) at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%, of the s in
the library are unique members (which encode matched AC element and BC elements
sequences); or
e) less than 20%, 15%, 10%, 5%, 4%, 3%, 2%, or 1%, of the members in the library
are unique members (which encode matched AC element and BC elements sequences).
192. The library of paragraph 191, wherein each unique member of the plurality is
configured such that, when expressed, the AC element, e.g., the ACVRS, and the BC t, e.g.,
the BCVRS, form a functional antigen g molecule, e.g., a single chain TCR.
193. The library of any of paragraphs 191-192, wherein the library is a display library.
194. The library of any of aphs 191-193, wherein each of the members of the plurality
further encodes a polypeptide that results in display of the member on the surface of a display entity.
195. The library of any of paragraphs 191-194, wherein the library is a phage display library.
196. The library of any of paragraphs 191-194, wherein the library is a yeast display library.
197. The library of any of paragraphs 191-194, wherein the library is a mammalian display
library.
198. A method of making a binding polypeptide, the method comprising:
a) acquiring a library of any of paragraphs 191-197; and
b) expressing a polypeptide encoded by a unique nucleic acid of the library.
199. The method of paragraph 198, further comprising ting the polypeptide with an
antigen.
200. The method of paragraph 198 or 199, further comprising retrieving the nucleic acid that
encodes a polypeptide that binds the antigen.
201. A method of making a nucleic acid sequence comprising a sequence that encodes an 7
chain element (GC element) of a TCR 7 chain le region (GCVR) and a 5 chain element (DC
element) of a TCR 5 chain variable region (DCVR), and wherein the GCVR and DCVR are matched,
the method sing:
a) acquiring an isolated production on site, e.g., a production micro-chamber,
comprising:
i) a 7 chain (GC) strand, wherein the GC strand is a strand of a 7 chain double-
stranded cDNA (GC ds cDNA) comprising a segment that encodes a GC element of the
GCVR from a cell, e.g., a 7 chain variable region ce (GCVRS); and
ii) a 5 chain (DC) strand, wherein the DC strand is a strand of a 5 chain double-
stranded cDNA (DC ds cDNA) comprising a segment that encodes a DC element of the
DCVR from the cell, e.g., a 5 chain variable region sequence ), and
b) covalent g, e.g., ligation, of a GC strand to a DC ,
wherein the isolated production on site, e.g., a production micro-chamber, does not
include a nucleic acid encoding a DCVR or a GCVR from a cell other than the cell,
y making a nucleic acid sequence comprising a sequence that encodes a GC element of
a GCVR and a DC element of a DCVR, n the GCVR and DCVR are matched.
WO 19402
202. The method of paragraph 201, wherein the GC element comprises, or consists of, a
GCVRS, or a functional nt thereof (e. g., an antigen binding nt thereof).
203. The method of paragraph 201 or 202, wherein the DC element comprises, or consists of,
a DCVRS, or a functional nt thereof (e.g., an antigen binding fragment thereof).
204. The method of any of paragraphs 201-203, wherein the nucleic acid sequence is
configured such that, when expressed, the GCVRS and the DCVRS form a functional antigen binding
molecule, e. 3., a single chain TCR molecule.
205. The method of aph 204, wherein the antigen binding molecule is onal in
vitro, ex vivo, or in viva, e.g., as ined by a method or assay described herein.
206. The method of any of paragraphs 201-205, wherein acquiring an isolated production
reaction site, e.g., a production micro-chamber, comprises:
a) acquiring a capture substrate bound to:
(i) a first double-stranded cDNA (ds cDNA) sing a strand that is
mentary to a first mRNA that encodes a GCVR from a cell; and
(ii) a second ds cDNA comprising a strand complementary to a second mRNA
encoding a DCVR from the cell (the cDNA loaded capture substrate), and
b) maintaining the isolated production reaction site, e.g., the production micro-chamber,
under conditions that allow amplification of the first and second ds cDNAs, to produce:
a plurality of GC ds cDNAs comprising a segment that encodes a GC element of the GCVR
from the cell, e. g., a GCVRS; and
a plurality of DC ds cDNAs comprising a segment that encodes a DC element of the DCVR
from the cell, e.g., a DCVRS.
207. The method of paragraph 206, wherein the GC ds cDNA is identical, or ntially
identical, to the first ds cDNA.
208. The method of paragraph 206 or 207, wherein the DC ds cDNA is identical, or
substantially identical, to the second ds cDNA.
209. The method of any of paragraphs 206-208, wherein the capture substrate comprises a
bead, e.g., a magnetic bead.
210. The method of any of paragraphs 206-209, wherein the capture substrate comprises a
moiety (e.g., an oligonucleotide) which binds to cDNA, e.g., (i) a moiety which binds to the GC
strand; (ii) a moiety which binds to the DC strand; or (iii) both (i) and (ii).
211. The method of any of paragraphs 206-220, wherein the first mRNA and the second
mRNA are disposed on an mRNA loaded capture substrate.
212. The method of any of paragraphs 206-211, wherein the isolated production reaction site,
e.g., the production micro-chamber, comprises:
a reagent mixture suitable for producing, from the first and second mRNAs (e.g., after the
first and second mRNAs are released from the mRNA loaded e substrate into a solution), a first
ds cDNA sing a segment that encodes a GC element of the GCVR of the cell, e.g., a GCVRS,
and a second ds cDNA comprising a segment that encodes a DC element of the DCVR of the cell,
e.g., a DCVRS.
213. The method of any of paragraphs 206-212, wherein the isolated production on site,
e.g., production micro-chamber, comprises primers that mediate the production of the first ds cDNA.
214. The method of any of paragraphs 206-213, wherein the isolated production reaction site,
e.g., production micro-chamber, comprises primers that mediate the production of the second ds
cDNA.
215. The method of any of paragraphs 206-214, wherein a cDNA strand that is
complementary to a first mRNA that encodes a GCVR from a cell is made by reverse transcription of
the first mRNA.
216. The method of any of paragraphs 206-215, wherein a cDNA strand that is
mentary to a second mRNA that encodes a DCVR from a cell is made by reverse transcription
of the second mRNA.
217. The method of paragraph 215 or 216, n the reverse transcription takes place in
the isolated production reaction site, e.g., a tion-micro chamber.
218. The method of paragraph 215 or 216, wherein the reverse transcription takes place in an
isolated cell reaction site, e. g., a cell isolation micro-chamber.
219. The method of paragraph 215 or 216, wherein the reverse transcription takes place
outside the ed production reaction site, e.g., a production micro-chamber, or outside an isolated
cell reaction site, e.g., a cell ion micro-chamber.
220. The method of paragraph 215 or 216, wherein the reverse transcription takes place
outside the isolated production reaction site, e.g., a production-micro chamber, and outside an isolated
cell reaction site, e.g., a cell isolation chamber.
221. The method of paragraph 215 or 216, wherein the reverse transcription takes place
outside an isolated on site, e.g., outside a micro-chamber.
222. The method of any of paragraphs 206-221, wherein the amplification ses 20 or
fewer cycles, e.g., 25 or fewer, 24 or fewer, 23 or fewer, 22 or fewer, 21 or fewer, 20 or fewer, 9 or
fewer, 8 or fewer, 7 or fewer, 6 or fewer, or 5 or fewer cycles.
223. The method of any of paragraphs 206-222, wherein the reverse transcription and/or
amplification uses one or more primers, e.g., comprising a sequence specific for a GCVRS and/or a
DCVRS.
224. The method of any of paragraphs 206-223, wherein the amplification comprises using
two or more s that mediate the production of the GC ds cDNA, wherein at least one primer
ses a nucleotide ation, and wherein at least one primer does not comprise a nucleotide
modification.
225 . The method of any of paragraphs 206-224, wherein the amplification comprises using
two or more primers that mediate the production of the DC ds cDNA, wherein at least one primer
comprises a nucleotide modification, and wherein at least one primer does not comprise a nucleotide
modification.
226. The method of aph 225, wherein at least one primer comprises a nucleotide
modification, e.g., which reduces, e.g., inhibits, DNA synthesis, e.g., by a DNA polymerase.
227. The method of paragraph 225 or 226, wherein at least one primer does not comprise a
nucleotide modification, e.g., which s, e.g., inhibits, DNA synthesis, e.g., by a DNA
polymerase.
228. The method of paragraph 226 or 227, n the nucleotide ation inhibits a
DNA polymerase from extending the DNA.
229. The method of any of aphs 226-228, wherein the nucleotide modification is an
insertion of a spacer to the primer, e.g., between two adjacent nucleotides in the primer.
230. The method of paragraph 229, n the spacer is a flexible spacer, a carbon spacer
(e.g., -(CH2)n-, wherein n=3, 4, 5, or more), two or more (6.3., three, four, five, or more) abasic
nucleotides or a PEG spacer.
231. The method of any of aphs 226-228, wherein the nucleotide ation is 2’-O-
methyl, 2’-OH, 2’-NH2, or uracil, e.g., to a ribose.
232. The method of any of paragraphs 226-231, wherein the nucleotide modification is
located internally or at the 3’ end of the primer.
233. The method of any of paragraphs 223-232, wherein at least one primer comprises (i) a
first member; (ii) a second member; and optionally (iii) a nucleotide modification described herein,
e.g., located n (i) and (ii).
234. The method of paragraph 233, wherein the first member is capable of annealing with the
second member in the same primer or a different primer, e.g., forming a hairpin structure (Via
intramolecular ization) or a double-stranded structure (Via intermolecular ization)
sing a duplex region of 4, 5, 6, 7, 8, 9, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 20, more
basepairs.
235 . The method of paragraph 233 or 234, wherein the first member comprises a sequence
that is complementary to the sequence of an oligonucleotide ed to the capture substrate.
236. The method of any of paragraphs 233-235, wherein the second member comprises (6g,
from 5 ’ to 3’) one, two, or all of: (i) a sequence that is complementary to at least a portion of the first
member; (ii) a universal priming sequence (e.g., for PCR amplification or eneration
sequencing); and (iii) a sequence complementary to a target sequence, e.g., a GCVRS and/or a
DCVRS, optionally, wherein the second member comprises a sequence for homologous
recombination (e.g., in a yeast or mammalian cell).
237. The method of any of paragraphs 223-236, wherein at least one primer comprises a
sequence encoding at least a portion of a linker sequence, or a complementary sequence thereof.
238. The method of aph 237, n the primer that comprises a sequence encoding
at least a portion of a linker sequence, or a complementary sequence thereof, is phosphorylated, e.g.,
’ phosphorylated,
239. The method of paragraph 237 or 238, wherein the linker sequence ses, or consists
of, (Gly-Gly-Gly-Gly-Ser)n, where n=1, 2, 3, 4, 5, or more.
240. The method of any of paragraphs 9, wherein the GC ds cDNA comprises a 5’
overhang, e.g., a 5’ overhang that is e of hybridizing to an oligonucleotide ed to a capture
substrate.
241. The method of any of paragraphs 201-240, wherein the GC ds cDNA comprises a blunt
end, e.g., a blunt end comprising a 5’ phosphate.
242. The method of any of paragraphs 201-241, wherein the DC ds cDNA comprises a 5’
overhang, e.g., a 5’ overhang that is capable of hybridizing to an oligonucleotide attached to a capture
substrate.
243. The method of any of paragraphs 201-242, wherein the DC ds cDNA comprises a blunt
end, e.g., a blunt end comprising a 5’ phosphate.
244. The method of any of paragraphs 3, wherein the GC ds cDNA and the DC ds
cDNA comprise sticky ends, e.g., both have 5’ overhangs.
245. The method of any of paragraphs 4, wherein the GC strand and the DC strand are
covalently linked, e.g., ligated, to produce a single stranded nucleic acid sequence, wherein the GC
and DC strands are both sense s or both antisense strands.
246. The method of any of paragraphs 201-245, wherein a denatured GC strand of the GC ds
cDNA to a denatured DC strand of the DC ds cDNA are covalently , e.g., ligated, wherein the
GC and DC strands are both sense strands or both antisense strands.
247. The method of any of paragraphs 201-245, wherein the GC strand is present in the GC
ds cDNA and the DC strand is present in the DC ds cDNA, and wherein the GC ds cDNA and the DC
ds cDNA are covalently , e.g., ligated, e.g., to produce a double stranded nucleic acid sequence.
248. The method of any of paragraphs 201-247, wherein the covalent linking, e. g., ligation,
occurs in the isolated production reaction site.
249. The method of paragraph 248, wherein the isolated production reaction site, e.g., a
production micro-chamber, comprises a t that is capable of covalently linking, e.g., ligating, the
GC and DC strands or the GC and DC ds cDNAs.
250. The method of paragraph 248 or 249, wherein the ed production on site, e.g.,
a production micro-chamber comprises an enzyme that covalently couples the GC and DC strands or
the GC and DC ds cDNAs.
251. The method of any of paragraphs 201-247, wherein the covalent linking, e.g., ligation,
occurs in a site different from the isolated production reaction site, e.g., occurs in an isolated linkage
reaction site, e.g., a linkage micro-chamber.
252. The method of aph 251, n the GC strand and the DC strand are transferred
from the isolated production site to the isolated linkage reaction site, e. g., a linkage micro-chamber,
and the covalent linking occurs in the isolated e reaction site, e.g., a e micro-chamber.
253. The method of paragraph 251 or 252, n the isolated linkage reaction site, e.g., a
linkage chamber, comprises a reagent that is capable of covalently linking, e.g., ligating, the
GC and DC strands or the GC and DC ds cDNAs.
254. The method of any of paragraphs 25 1-25 3, wherein the ed linkage reaction site,
e.g., a linkage micro-chamber, comprises an enzyme that covalently couples the GC and DC s
or the GC and DC ds cDNAs.
255. The method of paragraph 250 or 254, wherein the enzyme is a , e.g., a l
stable ligase.
256. The method of any of paragraphs 251-255, wherein the covalent linking, e.g., ligation,
ses:
(a) heating the isolated linkage reaction site, e.g., the linkage micro-chamber, under
conditions (e.g., at 96°C) that allow denaturation of the GC strand and the DC strand;
(b) cooling the isolated linkage reaction site, e.g., the linkage micro-chamber, under
conditions (e.g., at 60-66°C) that allow hybridization of the splint oligonucleotide to the GC strand
and the DC strand;
(c) maintaining the isolated e reaction site, e.g., the linkage micro-chamber, under
conditions (e.g., at 46-66°C) that allow ligation of the GC strand and the DC strand (e.g., formation of
phosphodiester bond between the GC strand and the DC strand); and
(d) repeating steps (a), (b), and (c) sequentially for 2, 6, 20, 26, 20, 26, 30, 36, 40, 46, 60, or
more cycles.
257. The method of any of paragraphs 201-256, wherein the GC strand and the DC strand are
ntly linked, e.g., ligated, in the presence of a splint oligonucleotide.
258. The method of paragraph 257, wherein the splint oligonucleotide is hybridized to a
sequence comprising the junction of the GC strand and the DC strand, or a sequence complementary
thereof, and forms a duplexed region at the site of ligation.
259. The method of paragraph 257 or 258, wherein the splint oligonucleotide comprises a
modification (e.g., an NHZ group) that inhibits DNA synthesis, e.g., by a DNA polymerase.
260. The method of paragraph 259, wherein the modification is at the 3’ end of the splint
ucleotide.
261. The method of any of paragraphs 201-260, wherein a strand complimentary to the
covalently , e. g., ligated, GC and DC strands is produced by amplification.
262. The method of any of paragraphs 201-261, further comprising, prior to acquiring the
isolated tion reaction site, e.g., a production micro-chamber, acquiring an mRNA loaded
capture substrate.
263. The method of paragraph 262, wherein acquiring the mRNA loaded capture substrate
comprising:
a) acquiring an isolated cell reaction site, e.g., a cell isolation micro-chamber, comprising:
i) a cell; and
ii) a capture substrate capable of binding a first mRNA encoding a GCVR from the
cell and a second mRNA encoding a DCVR from the cell; and
b) maintaining the isolated cell reaction site, e.g., the cell isolation micro-chamber, under
conditions that allow lysis of the cell and g of the capture ate with the first mRNA and the
second mRNA to form the mRNA loaded capture substrate,
wherein the isolated cell reaction site, e.g., cell isolation micro-chamber, does not include a
nucleic acid encoding a GCVR or a DCVR from a cell other than the cell.
264. The method of paragraph 263, wherein the isolated cell reaction site, e.g., cell ion
micro-chamber, comprises a lysing reagent, e.g., a detergent.
265. The method of paragraph 263 or 264, n the cell is lysed by heat or an enzyme.
266. The method of any of paragraphs 263-265, wherein the capture substrate comprises a
moiety (e.g., an oligonucleotide) which binds mRNA, e.g., an oligo(dT).
267. The method of any of paragraphs 262-266, further comprising releasing the mRNA
loaded capture substrate from the isolated cell reaction site, e.g., the cell isolation micro-chamber.
268. The method of paragraph 267, wherein the releasing step is med in the presence of
a poly(dA) or poly(dT) oligonucleotide, e.g., to reduce cross-binding of non-captured mRNA.
269. The method of paragraph 262-268, wherein the mRNA loaded capture substrate is
transferred from the isolated cell reaction site, e.g., the cell isolation micro-chamber, to the isolated
production reaction site, e.g., the production micro-chamber.
270. The method of any of paragraphs 201-269, sing releasing the nucleic acid
sequence from the production chamber.
271. The method of any of aphs 201-270, further comprising amplifying the c
acid sequence.
272. The method of any of paragraphs 201-271, comprising cing all or a portion of the
nucleic acid sequence.
273. The method of any of paragraphs 201-272, comprising inserting all or a portion of
c acid sequence into a vector.
274. The method of aph 273, wherein the vector supplies an additional GC element or
DC element not included in the nucleic acid sequence.
275. The method of paragraph 273 or 274, wherein the vector supplies a GC CDRl, a GC
CDR2, or both.
276. The method of any of paragraphs 273-275, comprising expressing the .
277. The method of any of paragraphs 201-276, comprising expressing the nucleic acid
sequence to produce a polypeptide comprising a segment that encodes a GC element of the GCVR,
e.g., a GCVRS, and a segment that encodes a DC element of the DCVR, e.g., a DCVRS.
278. The method of paragraph 277, wherein the DC element is N—terminal to the GC element
in the polypeptide.
279. The method of paragraph 277, wherein the GC element is C-terminal to the DC element
in the polypeptide.
280. The method of any of paragraphs 277-279, further comprising contacting the
polypeptide with an antigen.
281. The method of any of paragraphs 277-280, r sing determining if the
ptide binds the n.
282. The method of any of paragraphs 201-281, wherein the cell is an immune cell, e.g., a B
cell or T cell, e.g., a human B cell or T cell.
283. The method of any of aphs 2, wherein the cell is a mammalian cell or an
avian cell.
284. A method of making a nucleic acid sequence comprising a sequence that encodes a 7
chain element (GC element) of a TCR y chain variable region (GCVR) and a 5 chain element (DC
element) of a TCR 5 chain variable region , and wherein the GCVR and DCVR are matched,
comprising:
a) acquiring an isolated cell reaction site, e.g., a cell isolation micro-chamber, comprising:
i) a cell; and
ii) a capture substrate capable of binding a first mRNA encoding a GCVR from the
cell and a second mRNA encoding a DCVR from the cell;
b) maintaining isolated cell on site, e.g., the cell isolation micro-chamber, under
conditions that allow lysis of the cell and binding of the capture substrate with the first mRNA and the
second mRNA to form the mRNA loaded capture substrate,
wherein the isolated cell reaction site, e.g., cell isolation micro-chamber, does not include a
c acid encoding a GCVR or a DCVR from a cell other than the cell;
c) contacting the mRNA loaded capture substrate with a reaction mixture, e.g., a reaction
mixture comprising reverse transcriptase, that uses the loaded mRNA as a template to make cDNA
(this can occur, e.g., in the isolated cell reaction site, in the isolated tion reaction site, or in
r, e. g., not in an isolated reaction site);
d) acquiring an isolated production reaction site, e.g., a production chamber,
comprising:
i) an y chain (GC) strand from step b), wherein the GC strand is a strand of a 7 chain
double-stranded cDNA (GC ds cDNA) comprising a segment that encodes a GC element of
the GCVR from the cell, e.g., a 7 chain variable region sequence (GCVRS); and
ii) a 5 chain (DC) strand from step b), wherein the DC strand is a strand of a 5 chain
double-stranded cDNA (DC ds cDNA) comprising a segment that encodes a DC t of
the DCVR from the cell, e.g., a 5 chain variable region sequence ); and
e) nt linking, e. g., ligation, of a GC strand to a DC strand,
wherein the ed production reaction site, e.g., a tion micro-chamber, does not
include a nucleic acid encoding a DCVR or a GCVR from a cell other than the cell.
285. A method of making a nucleic acid sequence comprising a sequence that s a y
chain element (GC element) of a TCR 7 chain variable region (GCVR) and a 5 chain element (DC
element) of a TCR 5 chain variable region (DCVR), and wherein the GCVR and DCVR are matched,
comprising:
a) acquiring an isolated cell reaction site, e.g., a cell isolation micro-chamber, comprising:
i) a cell; and
ii) a capture substrate capable of binding a first mRNA encoding a GCVR from the
cell and a second mRNA encoding a DCVR from the cell;
b) maintaining the isolated cell reaction site, e.g., the cell ion micro-chamber, under
conditions that allow lysis of the cell and binding of the capture substrate with the first mRNA and the
second mRNA to form the mRNA loaded capture substrate,
wherein the isolated cell reaction site, e.g., cell isolation micro-chamber, does not include a
nucleic acid ng a GCVR or a DCVR from a cell other than the cell;
c) ing an isolated production reaction site, e.g., a production micro-chamber, comprises:
ting the mRNA loaded capture substrate with a reaction mixture, e.g., a reaction mixture
comprising reverse transcriptase, that uses the loaded mRNA as a template, to produce:
a first double-stranded cDNA (ds cDNA) comprising a strand that is complementary to a first
mRNA that encodes a GCVR from a cell; and
a second ds cDNA comprising a strand complementary to a second mRNA encoding a DCVR
from the cell (the cDNA loaded capture substrate);
wherein the isolated production reaction site, e.g., a tion micro-chamber, does not
include a nucleic acid encoding a DCVR or a GCVR from a cell other than the cell.
d) maintaining the isolated production reaction site, e.g., the production micro-chamber,
under conditions that allow amplification of the first and second ds cDNAs, to produce:
a plurality of GC ds cDNAs comprising a segment that s a GC element of the GCVR
from the cell, e. g., a GCVRS; and
a plurality of DC ds cDNAs comprising a segment that encodes a DC element of the DCVR
from the cell, e.g., a DCVRS;
e) acquiring an isolated linkage reaction site, e.g., a linkage micro-chamber, comprising:
covalent linking, e.g., ligation, of a strand of the GC ds cDNA (GC strand) to a strand of the DC ds
cDNA (DC ), wherein the GC and DC strands are both sense strands or antisense strands; and
f) amplifying the covalently , e.g., ligated, GC and DC strands.
286. A method of making a y comprising a plurality of unique members, the method
comprising:
making the plurality of members, wherein each of the members comprises a sequence that
encodes a 7 chain element (GC element) of a 7 chain variable region (GCVR) and a 5 chain element
(DC element) of a 5 chain variable region (DCVR), and wherein the GCVR and DCVR are d,
made by a method of any of paragraphs 201-285,
n each unique nucleic acid sequence of the plurality comprises a GC element and a DC
element from a different unique cell,
thereby making a library comprising a plurality of unique s.
287. The method of paragraph 86, wherein the plurality of unique members comprises at
least 204, 205, 206, 207, 208, or 209 unique members.
288. The method of paragraph 286 or 287, wherein the plurality of unique members
comprises 204 to 209, 204 to 208, 204 to 207, 204 to 206, 204 to 205, 208 to 209, 207 to 209, 206 to 209, 205
to 209, 205 to 208, 206 to 207, 204 to 205, 205 to 206, 206 to 207, 207 to 208, or 208 to 209 unique
members.
289. The method of any of paragraphs 286-288, n at least 80%, 85%, 90%, 95%, 96%,
97%, 98%, 99%, or 200%, of the members in the library are unique members (which encode matched
GC element and DC elements sequences).
290. The method of any of paragraphs 286-289, wherein less than 20%, 25%, 20%, 5%, 4%,
3%, 2%, or 1%, of the members in the library are unique members (which encode d GC
element and DC elements ces).
291. A library comprising:
a plurality of unique members,
wherein,
i) each unique member of the plurality comprises a segment that s a GC element, e.g.,
a GCVRS, and a segment that s a DC element, e.g., a DCVRS, wherein the GC element and
the DC element in each unique member is matched;
ii) each unique member of the plurality ses a segment that encodes a GC element, e. g.,
a GCVRS, and a segment that encodes a DC element, e.g., a DCVRS, from a different unique cell;
iii) the library comprises one or more (e.g., two, three, four, or all) of the following
properties:
a) the library is made by a method of any of paragraphs 201-285;
WO 19402
b) the plurality of unique members comprises at least 204, 205, 206, 207, 208, or 209
unique nucleic acid sequences;
c) the plurality of unique members comprises 204 to 209, 204 to 208, 204 to 207, 204 to
206, 204 to 205, 208 to 209, 207 to 209, 206 to 209, 205 to 209, 205 to 208, 206 to 207, 204 to 205,
205 to 206, 206 to 207, 207 to 208, or 208 to 209 unique members;
d) at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 200%, of the members in
the library are unique members (which encode matched GC element and DC elements
sequences); or
e) less than 20%, 25%, 20%, 5%, 4%, 3%, 2%, or 1%, of the members in the library
are unique members (which encode matched GC element and DC elements sequences).
292. The library of paragraph 291, wherein each unique member of the plurality is
configured such that, when expressed, the GC element, e.g., the GCVRS, and the DC t, e.g.,
the DCVRS, form a functional antigen binding molecule, e.g., a single chain TCR.
293. The library of any of paragraphs 291-292, wherein the library is a display library.
294. The y of any of paragraphs 291-293, wherein each of the members of the ity
further encodes a polypeptide that results in display of the member on the surface of a display entity.
295. The library of any of paragraphs 291-294, wherein the library is a phage display library.
296. The library of any of paragraphs 4, wherein the library is a yeast display library.
297. The library of any of paragraphs 291-294, wherein the library is a mammalian display
library.
298. A method of making a binding polypeptide, the method comprising:
a) acquiring a library of any of paragraphs 291-297; and
b) expressing a polypeptide encoded by a unique nucleic acid of the library.
299. The method of paragraph 298, r comprising contacting the polypeptide with an
n.
300. The method of paragraph 298 or 299, further sing ving the nucleic acid that
encodes a ptide that binds the antigen.
301. An isolated production reaction site, e.g., a production micro-chamber, which is an
isolated production reaction site described in any of paragraphs l-85, 5, or 5.
302. The isolated production reaction site, e.g., a production micro-chamber, of paragraph
401, which does not e a nucleic acid encoding (i) an HCVR or an LCVR, (ii) an ACVR or a
BCVR, or (iii) a GCVR or a DCVR, from a cell other than the cell,
303. The isolated production reaction site, e.g., a production micro-chamber, of paragraph
301 or 302, which comprises one, two, or all of:
(i) one or more primers specific to V gene sequences of the (a) HC and LC, (b) a chain and [3
chain, or (c) 7 chain and 5 chain;
(ii) one or more primers specific to overhangs introduced onto the (a) HC and LC, (b) or chain
and [3 chain, or (c) 7 chain and 6 chain, cDNAs;
(iii) one or more primers comprising a first member, a second member, and a nucleotide
modification (e.g., a spacer) located between the first and second members, n the first member
is capable of annealing with the second member in the same primer or a different primer, e. g., forming
a hairpin structure (via olecular hybridization) or a double-stranded structure (via
intermolecular hybridization) comprising a duplex region of 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16,
17, 18, 19, 20, more basepairs.
304. The isolated production reaction site, e.g., a production micro-chamber, of any of
paragraphs 301-103, which does not comprise a reagent that can covalently link nucleic acids, e.g., a
ligase, e.g., a thermostable ligase.
305. The isolated production reaction site, e.g., a production micro-chamber, of any of
paragraphs 3, which comprises a reagent that can covalently link nucleic acids, e.g., a ligase,
e. g., a thermostable .
306. A self-annealing oligonucleotide comprising a first member, a second member, and a
nucleotide modification (e.g., a spacer) located between the first and second members, wherein the
first member is capable of annealing with the second member in the same oligonucleotide or a
different oligonucleotide, e. g., forming a hairpin structure (via intramolecular hybridization) or a
double-stranded structure (via intermolecular hybridization) sing a duplex region of 4, 5, 6, 7,
8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, more basepairs.
307. The oligonucleotide of paragraph 306, wherein the first and second members are
capable of forming a hairpin structure sing a duplex region of 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14,
, 16, 17, 18, 19,20, more basepairs.
308. The ucleotide of paragraph 306 or 307, wherein the first member is 5-40
nucleotides, e.g., 5-10, 5-20, 5-30, 30-40, 20-40, 10-30, 10-30, or 15-25 nucleotides, in length.
309. The oligonucleotide of any of paragraphs 8, wherein the second member is 5-40
nucleotides, e.g., 5-10, 5-20, 5-30, 30-40, 20-40, 10-30, 10-30, or 15-25 nucleotides, in length.
310. The oligonucleotide of any of paragraphs 306-309, wherein the spacer is a flexible
spacer or a PEG spacer.
311. The oligonucleotide of any of paragraphs 306-310, n the first member ses
a ce that is complementary to the sequence of an oligonucleotide attached to a capture
ate.
312. The oligonucleotide of any of aphs 306-311, wherein the second member
comprises (e.g., from 5’ to 3’) one, two, or all of: (i) a sequence that is complementary to at least a
portion of the first member; (ii) a universal priming sequence (e.g., for PCR amplification or next-
generation sequencing); and (iii) a sequence mentary to a target sequence, e.g., a GCVRS
andfor a DCVRS, optionally, wherein the second member comprises a sequence for homologous
recombination (e.g., in a yeast or mammalian cell).
313. An isolated linkage reaction site, e. g., a linkage micro-chamber, which is an isolated
linkage reaction site described in any of paragraphs 51-83, 85, 151-183, 185, 251-283, or 285.
314. The isolated linkage reaction site, e.g., a linkage micro-chamber, of paragraph 313,
which does not include a c acid encoding (i) an HCVR or an LCVR, (ii) an ACVR or a BCVR,
or (iii) a GCVR or a DCVR, from a cell other than the cell,
315. The isolated linkage reaction site, e.g., a linkage micro-chamber, of aph 313 or
314, which comprises a splint ucleotide that is capable of hybridizing to a sequence comprising
the on of (i) the HC strand and the LC strand, (ii) the AC strand and the BC strand, or (iii) the
GC strand and the DC strand, or a sequence complementary thereof, to form a duplexed region at the
site of ligation.
316. The isolated linkage reaction site, e.g., a linkage chamber, of any of paragraphs
313-315, which comprises a t that can covalently link nucleic acids, e.g., a ligase, e.g., a
thermostable .
317. The method of any of paragraphs 1-300, which does not include a step of p
extension rase chain reaction (OE-PCR), also known as splicing by overlap extension or
splicing by overhang extension (SOE) PCR (Higuchi er al., Nucleic Acids Res. 1988; :7351-
67), e.g., in the linking step.
EXAMPLES
Example 1: Cohesive-End PCR-Ligation in Drops
B cells were encapsulated in droplets. B cells were encapsulated into droplets, with droplet
volume ranging from 10 pL to 100 nL, typically ~100-1000 pL. Sources of B cells can include, for
example, mice, human, rat, rabbit, or chicken. The carrier (oil) phase was composed of 3M HFE-
7500 with ~1% fluorosurfactant (RAN Biotechnologies). Droplets were formed by a microfluidic
chip (e.g., 2R 100 pm from Dolomite) with flow of fluid phases controlled by a syringe or pressure
pump. The aqueous phase of droplets was composed of a buffer (e.g., Tris, pH 7.5), a detergent to aid
cell lysis, and magnetic beads which contain oligonucleotides (primers) to anneal to heavy and light
chain mRNAs. Occupancy of drops should be not more than 1 cell per droplet, and at least one bead
per droplet.
The ts were incubated to facilitate cell lysis. The emulsion was heated to improved
lysis efficiency in the presence of certain detergents (e.g., Tween 20). For example, the emulsion can
be heated to a temperature of 40°C, 50°C, 60°C, 70°C, or 80°C, for imately 5-60 minutes.
After the cells were lysed, mRNA was released and captured on beads by ing to
oligonucleotides on the beads.
The emulsion was broken and the beads were recovered. Emulsions (or coalesce different
solution phases) were broken using drop destabilizing reagent, such as PFO orooctanol). The
aqueous phase containing the beads was recovered. The beads were isolated using magnet. The
beads were washed and resuspended in a buffer (e.g., Tris, pH 7.5).
Reverse transcription (RT) was performed to create cDNA-beads (in a non-emulsion
reaction). The beads were resuspended in a buffer-enzyme mix to facilitate RT (e.g., Superscript II
RT). The reaction was incubated at 40°C for 15 minutes to facilitate RT. The beads were washed
once with a buffer (6.3., Tris, pH 7.5).
The recovered beads can be encapsulated for PCR-Ligation reaction. Droplets ranging in
volume from about 5 pL-500 pL, most commonly about 10-50 pL, can be used. The carrier (oil)
phase can be ed of 3M HFE-7500 ning ~2% fluorosurfactant (RAN hnologies).
Drops can be encapsulated with: beads which have cDNA; PCR reagents (including, e.g., a DNA
polymerase, e.g., Phusion® High-Fidelity DNA Polymerase (NEB), Q5® High-Fidelity DNA
Polymerase (NEB), Pfu DNA polymerase, KAPA DNA polymerase, Vent® DNA polymerase, or Taq
DNA polymerase); oligonucleotides that allow for amplification of VH and VL sequences; a
thermostable ligase (e.g., Taq DNA ligase, Pfu DNA ligase, Ampligase® thermostable DNA ligase,
Tsc DNA ligase, Rma DNA ligase, Tfi DNA ligase, or Tth DNA ); and optimized buffer
ions (compatibility to support both DNA polymerase and ligase enzymatic activities).
The scFV cassette was constructed as VL-Linker-VH, as tested in tubes. The order can be
switched (VH-Linker-VL) with no significant impact on function or performance. The reverse
primers of the VL sequence can contain an overhang sequence encoding a Linker sequence and with
at least 1 modified tide (e.g., 3 consecutive nucleotides with 2’-O-methyl modification). The
VL reverse primers can also contain 5’-phosphate red to be ate of ligase). The forward
s of the VH sequence can contain an overhang sequence encoding a Linker sequence and with
at least 1 modified nucleotide (e.g., 3 consecutive nucleotides with 2’-O-methyl modification). The
VH forward primers can also contain 5’-phosphate red to be substrate of ligase). The
occupancy of drops should be not more than 1 bead per drop.
Thermocycling can be performed with on. The generated emulsion can be transferred
to PCR tubes. Thermocycling can be performed using the following conditions: l denaturation at
95-98°C for 30 seconds to 2 minutes; 10-30 cycles of: denaturation at 95-98°C for 10-30 seconds,
primer annealing at 50-60°C for 10-30 seconds, rase extension at 72°C for 30 seconds, and
cohesive product annealing and ligation at 45-55°C for 3 minutes. The reaction can be hold at 4°C.
The emulsion was broken and the portion which contains linked (and non-linked) product was
recovered. ons can be broken using drop destabilizing reagent, such as PFO
(perfluorooctanol). The aqueous phase can be recovered and the beads can be discarded.
The linked product (representing natively linked VL—linker-VH), as tested in tubes, was
purified from non-linked VH and VL products. Ligated t was separated from non-ligated
products by size separation. For example, denaturing PAGE (polyacrylamide gel electrophoresis) or
denaturing HPLC-SEC can be used. Linked product (about 800-900 bp) was isolated from non-linked
t (about 350-500 bp). For denaturing PAGE purification, the ligated band was cut out from the
gel and electroelution was performed to extract DNA from the gel slice (Bio-Rad Electro-Elutor).
The purified linked product can be amplified by PCR. Polymerase/conditions which can
moderately read through DNA containing modified nucleotides, e.g., Taq polymerase, can be used.
Final PCR product can be introduced to yeast using standard methods (e.g., electroporation
with expression vector) to create a natively paired library derived from biological sources.
Example 2: Ligase Cycling ch
B cells were encapsulated in droplets. B cells were encapsulated into droplets, with droplet
volume g from 10 pL to 100 nL, typically ~100-1000 pL. Sources of B cells can include, for
example, mice, human, rat, rabbit, or chicken. The carrier (oil) phase was ed of 3M HFE-
7500 with ~1% fluorosurfactant (RAN Biotechnologies). Droplets were formed by a microfluidic
chip (e.g., 2R 100 pm from Dolomite) with flow of fluid phases controlled by a syringe or pressure
pump. The aqueous phase of ts was composed of a buffer (e.g., Tris, pH 7.5), a detergent to aid
cell lysis, and magnetic beads which contain oligonucleotides (primers) to anneal to heavy and light
chain mRNAs. Occupancy of drops should be not more than 1 cell per droplet, and at least one bead
per droplet.
The droplets were incubated to facilitate cell lysis. The emulsion was heated to improved
lysis efficiency in the presence of certain detergents (e.g., Tween 20). For example, the emulsion can
be heated to a temperature of 40°C, 50°C, 60°C, 70°C, or 80°C, for approximately 5-60 minutes.
After the cells were lysed, mRNA was released and captured on beads by annealing to
oligonucleotides on the beads.
The emulsion was broken and the beads were recovered. Emulsions (or coalesce different
on phases) were broken using drop ilizing t, such as PFO (perfluorooctanol). The
s phase ning the beads was recovered. The beads were isolated using magnet. The
beads were washed and resuspended in a buffer (6.3., Tris, pH 7.5).
Reverse transcription (RT) can be performed to create cDNA-beads (in a non-emulsion
reaction). The beads can be resuspended in a -enzyme mix to facilitate RT (e.g., Superscript 11
RT). The reaction can be incubated at 40°C for 15 minutes to facilitate RT. The beads can be washed
once with a buffer (e.g., Tris, pH 7.5).
Recovered beads were encapsulated for PCR reaction. Droplets ranging in volume from
about 5 pL—500 pL, most commonly about 10-50 pL, can be used. The carrier (oil) phase can be
composed of 3M HFE-7500 containing ~2% fluorosurfactant (RAN Biotechnologies). Drops can be
encapsulated with: beads which contain cDNA (some beads can be ‘empty’, which is not
problematic); PCR reagents ding, e.g., a DNA rase, e.g., Phusion® High-Fidelity DNA
Polymerase (NEB), Q5® High-Fidelity DNA rase (NEB), Pfu DNA polymerase, KAPA DNA
polymerase, Vent® DNA polymerase, or Taq DNA polymerase); ucleotides that allow for
amplification of VH and VL sequences.
The scFv cassette can be constructed as VL—Linker-VH. The order can be switched (VH-
Linker-VL) with no significant impact on on or performance. The VL reverse and VH forward
primers can contain an overhang sequence encoding Linker. The VH reverse primer can have a 5’-
phosphate. The occupancy of drops should be not more than 1 bead per drop.
Thermocycling can be performed with emulsion. The generated emulsion can be erred
to PCR tubes. The thermocycling can be med using standard PCR ions: initial
denaturation: 95-98°C for 30 seconds to 2 minutes; 10-30 cycles of: denaturation at 95-98°C for 10-
seconds, primer annealing at 50-60°C for 10-30 seconds, polymerase ion at 72°C for 30
seconds. The reaction can be hold at 4°C.
The emulsion can be broken and the beads can be recovered. Emulsions (or coalesce
different solution phases) can be broken using drop destabilizing reagent, such as PFO
(perfluorooctanol). The aqueous phase that contains beads can be recovered. The beads can be
isolated using magnet. The beads can be washed to remove PCR product not captured on beads. The
beads can be resuspended in a buffer (e.g., Tris, pH 7.5).
The recovered beads can be encapsulated for ligase cycling reaction. Droplets ranging in
volume from about 5 pL-SOO pL, most commonly about 10-50 pL, can be used. The carrier (oil)
phase can be composed of 3M HFE-7500 containing ~2% fiuorosurfactant (RAN Biotechnologies).
The drops can be ulated with a Stint oligonucleotide which is complementary and anneals to 3’
terminus of ‘top’ VL strain and 5’ terminus of ‘top’ VH strand; a thermostable ligase (e.g., Taq DNA
ligase, Pfu DNA ligase, Ampligase® thermostable DNA , Tsc DNA ligase, Rma DNA ligase,
Tfi DNA ligase, or Tth DNA ligase); reaction components required to support ligase tic
activity (e.g., NAD). The ncy should be not more than 1 bead per drop.
Thermocycling can be performed with emulsion. The ted emulsion can be transferred
to PCR tubes. Thermocycling can be performed using standard conditions: 3-15 cycles of:
denaturation: 90-95°C for 30 seconds, annealing and ligation at 50-60°C for 1-3 minutes. The
reaction can be hold at 4°C.
The emulsion can be broken and the aqueous portion which contains the linked product can
be recovered. Emulsions (or coalesce ent solution phases) can be broken using drop
destabilizing reagent, such as PFO (perfluorooctanol). The s phase can be recovered and the
beads can be discarded.
The d linked product can be amplified by PCR. Standard ions can be used with
oligonucleotides that anneal the outer termini of the linked VL-Linker-VH ligated fragment.
Final PCR product can be introduced to yeast to create a natively paired library derived from
biological sources.
Example 3: Ligase Cycling Reaction
In this example, VH and VL PCR products were covalently linked using a thermostable ligase
and a splint oligo.
RNA from a hybridoma clone (ATCC, HB-112, “4G2”) was isolated using an RNeasy Kit
(Qiagen). cDNA was generated using SuperScript IV Reverse Transcriptase (Thermo Fisher) and the
isolated RNA following manufacturer ended conditions. Primers used were directed to
constant regions of the heavy and light chains: mouse IgG_CHl_rev (5’-
CHGATGGGGSTGTYGTTKTRGC (SEQ ID NO: 1)) and mouse Kappa_Rev (5 ’-
GTGCAGCATCAGCCC (SEQ ID NO: 2)). As used herein, the tides are defined by IUPAC
nucleotide code, e.g., R=A or G; Y=C or T; S: G or C; K=G or T; H=A or C or T.
The components were mixed, heated to 65 °C for 5 min, and then incubated on ice for at least
1 min. The following components were then added:
ent Volume (111)
5X SSIV buffer 4
100mm
UT RNase Inhibitor (40 U/ul)
SuperScript IV Reverse Transcriptase
The components were mixed and then incubated at 55°C for 10 minutes. The on was
then inactivated by incubating at 80°C for 10 minutes. cDNA products were used as templates for
PCR to separately generate VH and VL products. The primers used included:
4G2 VL_Fwd
5’-GACATCAAGATGACCCAGTCTC (SEQ ID NO: 3)
Mouse Kappa_Rev-Phos
’-/5Phos/ACCAGCAGAGCTCTCACCTGGTGCAGCATCAGCCC (SEQ ID NO: 4)
4G2 VH_Fwd-Phos
hos/GGAACTACCGAAGGCACAGGTGAGGTCCAGCTGCAACAGTC (SEQ ID NO: 5)
Mouse IgG_CHl_rev
’-CHGATGGGGSTGTYGTTKTRGC (SEQ ID NO: 1)
WO 19402
The PCR reactions each included:
Q5 Hot Start 2X Master Mix (NEB)
Thermocycling was then performed as follows:
Initial
Denature Anneal Extend
Denaturation
Forever
PCR products were purified using AMPure beads. Purified PCR products were quantified by
Nanodrop. Ligase cycling reactions were set up in 25 ul reactions to achieve a 1:1:1 molar ratio of
VH product, VL product, and splint oligo. The splint oligo used had the following sequence:
TGTGCCTTCGGTAGTTCCACCAGCAGAGCTCTCACCTG/3AmMO/ (SEQ ID NO: 6).
Each ligase cycling reaction included:
Component Amount
4G2 VH PCR product 38 ng
4G2 VL PCR product 35 ng
Splint oligo (0.1 mM) 3.3 ul
Taq ligase buffer (NEB) 2.5 ul
Taq ligase (NEB) 1 ul
Water Up to 25 ul
The ligase cycling reactions were analyzed by denaturing PAGE (polyacrylamide gel
electrophoresis). In addition to Taq Ligase, an additional thermostable ligase gase (Amp),
Lucigen) was ted.
As shown in efficient g of VH and VL product was observed using both Taq
ligase and Ampligase thermostable ligase.
Example 4: Retention of Native Pairing During Bulk Re-Amplification of Linked Products
An emulsion OE-PCR workflow creates VH and VL ts which necessarily share
sequences to facilitate splicing by overlap extension. In such a workflow, any retained VH/VL
product which is not d together within a drop can become spliced during bulk (non-emulsion)
PCR ication of products. Since such splicing occurs during the bulk phase when all cell
products (e.g., many clones/unique sequences) are mixed, random (non-native) pairing could occur
(see Eur J l. 2013; 43(9):2507-15). The workflow would benefit from a strategy that not only
s this occurrence, but completely prevents it. Described herein is a method in which VH and
VL products do not share any common sequence, and thus the workflow is not susceptible to
ntal splicing by OE-PCR during bulk re-amplification steps.
To demonstrate this, linked VH-VL products were generated from two hybridoma clones
which differ from each other by size. Native versus tive pairing could therefore be assessed by
size of products. VH and VL products from hybridoma 4G2 were generated as described in Example
3. Hybridoma 9E10 (ATCC, CRL-l729) “mini” VH and VL ts were similarly produced using
the same RT primers and the following PCR primers:
9E10 Mini_VL_Fwd
’-GGCAGTGGGTCTGGGACAG (SEQ ID NO: 7)
mouse Kappa_ReV-Phos
5’-/5Phos/ACCAGCAGAGCTCTCACCTGGTGCAGCATCAGCCC (SEQ ID NO: 4)
9E10_VH_Fwd
’ -/5Phos/GGAACTACCGAAGGCACAGGTGAGGTGCACCTGGTGGAGTCTGGGGG (SEQ ID
NO: 8)
9E 10 Mini_VH_ReV
5’ —GGATAGTGGGTGTAAGTACCACGACTACCAATG (SEQ ID NO: 9)
The 4G2 VH and VL products produced were of size 400 bp and 378 bp, respectively. The
mini 9E10 VH and VL products produced were of size 203 bp and 168 bp, respectively. The PCR
ts were purified using AMPure beads.
The products were then subjected to ligase cycling reactions as described in Example 3, using
cycles of thermocycling. 4G2 and 9E10 VH-VL products were ligated in te tubes, simulating
generation of natively linked products in droplets (in individual compartments). The products were
analyzed by denaturing PAGE. Natively linked products of 4G2 VH-VL and of 9E10 VH-VL
corresponded to sizes of 778 bp and 371 bp, respectively. Twenty cycles of ligase cycling were used
to create products containing both ligated ts as well as non-ligated VH and VL DNA. As
shown in , natively linked VH-VL products were generated for both 6G2 and 9E10.
The ligation products were purified by AMPure beads and then combined and used as
template for PCR re-amplification of linked products. This step ted ry of products after
performing linking of VH-VL in drops, followed by PCR re-amplification of linked VH—VL. By
having non-ligated VH and VL DNA as template in the PCR, this provided an opportunity for DNA
to become linked together during bulk re-amplification by PCR. Since the DNA was not
compartmentalized by individual clones, any linkng that ed in bulk phase would lead to non-
native pairing.
PCR re-amplification was performed as described below. “Outer” primers used to amplify
linked products are as follows:
9E10 L_Fwd
’-GGCAGTGGGTCTGGGACAG (SEQ ID NO: 7)
4G2 VL_Fwd
’-GACATCAAGATGACCCAGTCTC (SEQ ID NO: 3)
9E 10 Mini_VH_Rev
5’-GGATAGTGGGTGTAAGTACCACGACTACCAATG (SEQ ID NO: 9)
Mouse l_rev
’-CHGATGGGGSTGTYGTTKTRGC (SEQ ID NO: 1)
The PCR reactions each included:
Q5 Hot Start 2X Master Mix
Fwd primers (10 uM) 0.9 (each)
Rev primers (10 uM) 0.9 (each)
Purified ligation products 4.5 (each)
Thermocycling was then performed as follows:
Initial
Denature Anneal Extend
Denaturation
Finally, products were ed by agarose gel electrophoresis. Non-natively linked ts
could be readily identified by their intermediate size. Specifically, 4G2-VL / 9E10-mini-VH and
9E10-mini-VL / 4G2 VH linked products would correspond to sizes of 581 bp and 568 bp,
WO 19402
respectively. As shown in , retention of native pairing was ed when mixed during bulk
re-amplification (lane 5).
Example 5: Oligo Design to Facilitate Capture of PCR Product onto Beads
After RT-PCR in droplets (e.g., as described above), the PCR product can be captured onto
beads within the drop to retain native matching of the VH and VL products. To facilitate efficient
capture of dsDNA PCR products onto beads, a strategy was devised to generate products which have
defined ssDNA at their ends. The sequence of this ssDNA is complementary to sequence of an oligo
conjugated to the beads. The complementarity of these sequences thus facilitates c and
ent capture of dsDNA PCR products.
PCR products from 4G2 cDNA were produced using PCR with the following primers and Q5
DNA polymerase. The reverse primer contains a 5’-biotin moiety to facilitate specific detection of
PCR product on beads. The PCR strategy was as follows:
(1) (VL) 4G2 VL Fwd +PEG Mus Kap Rev biotin
(2) (VL) 4G2 VL Fwd —PEG Mus Kap Rev biotin
(3) (VL) 4G2 VL Fwd No_5’ Mus Kap Rev biotin
(3) (VH) 4G2 VH Fwd Biotin Mus IgG-HC Rev +PEG
The following primers were used:
4G2 VL Fwd +PEG
= _
TGGATCGTTACTAATATTCGC/iSp18/GGACTCAGACACTTCCGTGCGACATCAAGATGACC
CAGTCTC (SEQ ID NO: 10)
4G2 VL Fwd -PEG
’ _
TGGATCGTTACTAATATTCGCGGACTCAGACACTTCCGTGCGACATCAAGATGACCCAGT
CTC (SEQ ID NO: 11)
4G2 VL Fwd No_5’
5’-GGACTCAGACACTTCCGTGCGACATCAAGATGACCCAGTCTC (SEQ ID NO: 12)
Mus Kap Rev biotin
iotinTEG/ACCAGCAGAGCTCTCACCTGGTGCAGCATCAGCCC (SEQ ID NO: 13)
Mus IgG-HC Rev +PEG
5a _
GCAATCCATCAACGTC/iSp l8/CGTGACACATGTGGTTCAAGTACGGCHGATGGGGSTGTY
GTTKTRGC (SEQ ID NO: 14)
4G2 VH Fwd Biotin
’-/5BiotinTEG/GGAACTACCGAAGGCACAGGTGAGGTCCAGCTGCAACAGTC (SEQ ID NO:
PCR products were analyzed by agarose gel electrophoresis, ed by AMPure beads and
quantified by Nanodrop.
Amine-labeled capture oligos were conjugated to carboxylated beads (COMPEL, Bangs
Laboratories) using standard methods. The following oligos having sequence complementary to
sequence 5’ of the PEG spacers in the above PCR primers were used for conjugation to beads. Beads
were conjugated to VL oligo only, to VH oligo only, or to both oligos.
VL Capture
’ _
GCGAATATTAGTAACGATCCAAAAAAAAAAAAAAAAAAAAA/iSp 18/iiSp1 8//iSp18//3AmM
O/ (SEQ ID NO: 16)
VH Capture
’ TGATGGATTGCAAAAAAAAAAAAAAAAAAAA/iSp18//iSp18//iSp18//3AmMO/
(SEQ ID NO: 17)
To capture PCR product, the following ons were setup. Purified PCR products were
d in 0.5X SSC buffer to achieve 250 ng DNA in 25 ul final volume. Oligo-conjugated beads
were washed with 0.5X SSC, and 400,000 beads were then mixed with each diluted PCR product. The
bead-PCR product mix was mixed by pipet to suspend the beads, and the tubes were then placed in a
thermocycler and the following m was run:
Step Temp Time
1 70 0C 30 sec
2 55 0C 30 sec
3 50 0C 30 sec
4 45 °C 4 min
40 OC 4 min
6 35 °C 4 min
7 4 OC hold
The samples were as follows:
Blank (no capture oligo) (1) (VL)
WO 19402
2 Blank (no capture oligo) (2) (VL)
3 Blank (no capture oligo) (3) (VL)
4 Blank (no capture oligo) (4) (VH)
VL capture (1) (VL)
6 VL capture (2) (VL)
7 VL capture (3) (VL)
8 VL capture (4) (VH)
9 VH capture (1) (VL)
VH capture (4) (VH)
11 VH and VL capture (1) (VL)
12 VH and VL capture (4) (VH)
After temperature incubation, the samples were applied to a magnet to collect beads on the
side of the tube. Supernatants were removed, and the beads were washed with 0.5X SSC buffer.
Each sample was probed for captured PCR product with 50 pl of AlexaFluor 647 IgG Fraction
Monoclonal Mouse Anti-Biotin antibody (Jackson) diluted 1:1000 in PBS containing 1% BSA. After
incubation with mixing for 25 minutes at room temperature, the tubes were applied to a magnet, the
supernatants were removed, and the beads were washed with PBS buffer. The beads were
ended in 50 ul PBS and then analyzed by flow cytometry.
As shown in FIGS. 5A-SB, the results demonstrate efficient and specific PCR product
capture by this method, with a requirement for a PEG spacer in the PCR primers and an appropriate
complementary oligo conjugated to the beads (e. g., as in sample (1)).
Example 6: Generation of Natively Paired VH-VL Product in Drops by Ligase g
In this e, cells expressing VH and VL sequences of antibody 4G2 were lysed and
mRNAs from the lysate were used to generate natively paired VH-VL products by ligase cycling.
Cell lysis buffer was prepared as s:
300 ul 20% Ficoll PM400
ul 20% Sarkosyl
40 ul 5 mM EDTA
100 ul 2M Tris pH 7.5
350 ul Water
200 ul 100% ep
Carboxylated COMPEL magnetic beads (Bangs Labs) were ated with the following
amine-modified oligos:
VL Capture
’ _
GCGAATATTAGTAACGATCCAAAAAAAAAAAAAAAAAAAAA/iSp 18//iSp1 8//iSp18//3AmM
O/ (SEQ ID NO: 16)
VH Capture
’ -GACGTTGATGGATTGCAAAAAAAAAAAAAAAAAAAA/iSp18//iSp18;’/iSp18//3AmMO/
(SEQ ID NO: 17)
Mus_IgG_CH1_mRNA capture
’ _
CTGGACAGGGATCCAKAGTTCCAAAAAAAAAAAAAAAAAAAAA/iSp18//iSp18//iSp18//3A
mMO/ (SEQ ID NO: 18)
Mus_Kap_mRNA capture
’ -GTGCAGCATCAGCCCGAAAAAAAAAAAAAAAAAAAA/iSp18//iSp18i/iSp18/l3AmMO/
(SEQ ID NO: 19)
Oligo(dT) may also be used for mRNA e (e.g., for capture of mRNAs encoding a VH
andfor VL).
Beads were suspended in lysis buffer at a density of 2x107 beads/ml, which facilitates
encapsulation of about 2.5 beads per drop with drop sizes used.
4G2 mouse hybridoma cells were washed with PBS and resuspended in PBS ning 0.1%
BSA and 24% Optiprep at a density of 2x106 cells/m1. This density facilitates encapsulation into drops
at imately one cell per two drops. For co-encapsulation of cells and beads in drops, a 2-
reagent, 100 pm diameter fluorophilic luidic chip (Dolomite) was used with solution flow
controlled by three Mitos P-Pump pressure pumps (Dolomite). The microfluidic chip ned two
input channels for aqueous solutions and two input ls for fluorocarbon oil. Cells and beads
were flowed at a rate of 10 til/min each, and fluorinated oil (HFE-7500 containing 1%
fluorosurfactant (RAN Biotechnologies)) was flowed at 55 til/min. These flow rates resulted in
droplets of approximately 500 pl in volume. Emulsions were collected for an hour.
The emulsions (cells and l control) were applied to a heat block at 45 0C for 20 min to
facilitate mRNA capture onto beads. The emulsions and beads were subsequently kept and handled at
4°C to reduce dissociation of mRNA from beads. Excess oil was removed by syringe, and 5 ml of
ice-cold 6X SSC buffer containing RNAse inhibitor (1:100) was added to the emulsion. PFO
(1H,1H,2H,2H-Perfluorooctanol) was added (100 ML) to the emulsion to induce drop coalescence.
The sample was mixed thoroughly by inversion followed by centrifugation at 500xg for 5 min at 4°C.
The aqueous phase was mixed by pipet, collected and then transferred to a new tube. After ng
the tube to the magnet, the beads (having hybridized mRNA) were washed twice with 500 ul of ice-
cold buffer (100 mM Tris HCl pH 8, 500 mM KCl, 15 mM MgC12) containing RNase inhibitor
(1 : 100). The beads were then suspended in 500 u] of ice-cold buffer (100 mM Tris HCl pH 8, 500
mM KCl, 15 mM MgC12) containing RNase inhibitor (1:100).
A second emulsion was prepared with the mRNA beads and RT-PCR mix. The mRNA beads
were suspended in the RT-PCR bead buffer (100 ul 2X RT—PCR buffer (OneTaq One-Step RT-PCR,
NEB), 70 ul of Optiprep, 4 ul of RNase inhibitor, 6.4 ul of ed 25 uM primers, and 32 ul of
water). Primers used were:
4G2 VL Fwd +PEG
’ _
TGGATCGTTACTAATATTCGC/iSp18/GGACTCAGACACTTCCGTGCGACATCAAGATGACC
CAGTCTC (SEQ ID NO: 10)
Mus IgG-HC Rev +PEG
’ _
CATCAACGTC/iSp18/CGTGACACATGTGGTTCAAGTACGGCHGATGGGGSTGTY
GTTKTRGC (SEQ ID NO: 14)
mouse Kappa_Rev-Phos
’—/5Phos/ACCAGCAGAGCTCTCACCTGGTGCAGCATCAGCCC (SEQ ID NO: 4)
4G2 VH Fwd-Phos
’-/5Phos/GGAACTACCGAAGGCACAGGTGAGGTCCAGCTGCAACAGTC (SEQ ID NO: 5)
Separately, enzyme mix was prepared (400 ul 2X RT-PCR buffer, 43 ul of enzyme diluted to
1.33X in RT-PCR buffer, 16 ul RNase inhibitor, and 341 ul water).
Emulsions for RT-PCR were generated by co-flowing beads and enzyme mix at 2.5 til/min
and 7.5 ul/min, respectively, and oil (HFE7500 containing 2% fluorosurfactant) at 35 ul/min. A two-
reagent 30 um diameter fluorophilic microfluidic chip (Dolomite) was used with pressure pumps to
generate droplets. Under these conditions, droplets of approximately 15-35 pl in volume were
, with bead occupancy of less than one bead per 5 drops. Drops were ted until all beads
were encapsulated (about 30-60 s). The emulsions were then aliquoted into PCR tubes for
thermocyling in a thermocycler with the following program:
Denature
1 Cycle
55°C 50°C 45°C 40°C 12°C
30 4 min Forever
sec sec
The thermocycled emulsions were combined into one tube. Excess oil was removed from the
bottom using a syringe. To break the drops, 100 ul of PFO was added followed by mixing and
centrifugation at 2000xg for two minutes. The aqueous phase was then mixed by pipet (to suspend
beads) and transferred to a new tube. The beads contained captured PCR product. The tube was
d to a magnet and supernatant removed. The beads were gently washed with 500 pl of ice-cold
0.5X SSC buffer. The beads were again applied to a magnet, supernatant removed and then
resuspnded in 100 pl ice-cold 0.5X SSC buffer.
Samples were then ed for ligase cycling emulsion to ntly link VH and VL
products. The beads were suspended in bead on (20 ul 10X Taq Ligase buffer (NEB), 117 pl
60% sucrose, and 63 ul water). Taq ligase mixture was prepared (80 ul 10X Taq Ligase buffer, 42 ul
Taq Ligase, 13 ul of splint oligo (at 0.1 uM), and 665 pl water). The splint oligo used was
’ —ACCTGTGCCTTCGGTAGTTCCACCAGCAGAGCTCTCACCTG/3AmMO/ (SEQ ID NO: 6).
Emulsions were generated by flowing the bead solution at 2 ul/min, enzyme mix at 6 ul/min,
and oil (HFE7500 containing 2% urfactant) at 35 ul/min. A two-reagent 30 um diameter
fluorophilic microfluidic chip (Dolomite) was used with pressure pumps to generate droplets. Under
these conditions, droplets of approximately 15-35 pl in volume were formed, with bead occupancy of
less than one bead per 5 drops. Drops were generated until all beads were encapsulated (about 30-60
s). The emulsion was then aliquoted into PCR tubes for thermocyling in a thermocycler using
the following program:
Initial Denature Anneal
Denaturation
1 cycle 40 cycles
2 minutes 20 30 1 Forever
seconds seconds minute
The thermocycled emulsions were ed into one tube. Excess oil was removed from the
bottom using a e. To break drops, 100 pl of PFO was added followed by mixing and
centrifugation at 2000xg for two minutes. The aqueous phase was then mixed by pipet (to suspend
beads) and transferred to PCR tubes (50 ul per tube). The samples were then applied to a thermocyler
preheated to 80°C to facilitate dissociation of ligated PCR products hybridized to beads. After
allowing beads to settle, the supernatant was removed and transferred to a new tube. The products
were then purified using AMPure beads.
The ligated products were then reamplified in tubes by PCR using the ing s:
—Fwd-Reamp 5’—TGACCCAGTCTCCATCTTCA (SEQ ID NO: 23)
4G2-VH-Rev-Reamp 5’—TGTTGTTTTGGCTGAGGAGA (SEQ ID NO: 24)
Component Volume (111)
Thermocycling was med as follows:
Initial
Denature Anneal Extend
Denaturation
The reamplification products were analyzed by agarose gel electrophoresis. As shown in the 4G2 cell sample yielded the intended t of linked VH-VL DNA, whereas negative control
samples did not yield any product.
It is contemplated that the final PCR product, containing, e.g., natively paired VL and VH
with a flexible linker sequence between in an intact open reading frame, can be cloned into a yeast
surface sion vector and transformed into yeast using standard methods, resulting in a natively
paired yeast y library. For example, an additional 50 bp on each terminus can be incorporated
by PCR. These 50 bp match appropriate sequences in a yeast expression vector to facilitate cloning
by homologous recombination in yeast, following yeast co-transformation of insert PCR product (e.g.,
containing natively linked VL-VH) and a linearized yeast expression vector.
Example 7: Self-Annealing Primers to Prevent PCR Product Capture ition from Primers
PCR product capture onto beads was tated by generation of sticky ends (ssDNA) on a
terminus of PCR products through incorporation of a spacer, such as PEG. These ssDNA portions
had sequence complementarity to oligos conjugated to beads, thereby facilitating specific
hybridization. The sequences in the PCR products were incorporated by their inclusion in
amplification primers. Since the PCR primers had the same sequences as the PCR product ssDNA
termini, they too can be captured onto beads h specific hybridization. Such primer capture
would necessarily e with PCR product capture.
Amplification of antibody repertoires by PCR typically requires a ude of PCR primers
to amplify the varied antibody gene ces. For e, primer sets recognizing V and J regions
of human and mouse repertoires are often composed of about 15-20 primers ed to the V regions
of the heavy and light . The requirement for a multitude of primers leads to circumstances that
exacerbate the ing effect of primers on capturing PCR product onto beads. For multiplex PCR
scenarios, in a given drop (compartment), a single primer (or limited ) can have
complementarity to the target antibody sequence within in the drop. The remaining repertoire primers
will not match the target sequence. These non-matching primers would not be incorporated into
amplicons during PCR, and as such, are poised to effectively compete for PCR product capture. To
address this tion, a design was ted that mitigates the ability of the unused PCR primers
within a drop to compete with PCR product for capture. PCR product capture can be attainable, for
example, under conditions of 15:1 molar ratio of unused primer to primer that generates amplicon.
Carboxylated ic beads were ated with the ing amine-modified oligos, as
described above.
VL_Capture
GCGAATATTAGTAACGATCCAAAAAAAAAAAAAAAAAAAAA/iSp l8//iSp l 8//iSp l8//3AmM
O/ (SEQ ID NO: 16)
VH_Capture
GACGTTGATGGATTGCAAAAAAAAAAAAAAAAAAAA/iSp lSli’iSp p18//3AmMO/ (SEQ
ID NO: 17)
G_CH1_mRNA_capt
CTGGACAGGGATCCAKAGTTCCAAAAAAAAAAAAAAAAAAAAA/iSp18//iSpl8//iSpl8//3A
mMO/ (SEQ ID NO: 18)
Mus_Kap_mRNA_capt
GTGCAGCATCAGCCCGAAAAAAAAAAAAAAAAAAAA/iSp18/!iSp18//iSpl8//3AmMO/ (SEQ
ID NO: 19)
RT-PCR reactions were set up to amplify VL sequence corresponding to 4G2 hybridoma
using the following oligos. These oligos represent a design without a self-annealing sequence (Non-
SA) and a design with a self-annealing sequence (SA). Additionally, an oligo that has a self-annealing
’ -end but has
sequence on its 5 a 3’ -end sequence that does not match the 4G2 VL or VH mRNA
sequences (Mismatch) was included as an exemplary PCR primer that would not be consumed during
PCR, akin to multiplex repertoire primers. The reverse primer was biotinylated to facilitate detection
of captured PCR product on the bead.
4G2_VL_Fwd_Non-SA
’ _
TGGATCGTTACTAATATTCGC/iSp18fGGACTCAGACACTTCCGTGCGACATCAAGATGACC
CAGTCTC (SEQ ID NO: 10)
Mismatch_VL_Fwd_Non-SA
WO 19402
’ _
TGGATCGTTACTAATATTCGC/iSp 18/GGACTCAGACACTTCCGTGCATTGTGCTGACGCAA
ACTGTTA (SEQ ID NO: 20)
4G2_VL_Fwd_SA
’ _
TGGATCGTTACTAATATTCGC/iSp 18/GAATATTAGTAACGATCCAGGACTCGGACCGACAT
CAAGATGACCCAGTCTC (SEQ ID NO: 21)
Mismatch_VL_Fwd_SA
’ _
GTTACTAATATTCGC/iSp 18/GAATATTAGTAACGATCCAGGACTCGGACCATTGT
GCTGACGCAAACTGTTA (SEQ ID NO: 22)
Mus_Kappa_ReV_Biotin
’-/5BiotinTEG/ACCAGCAGAGCTCTCACCTGGTGCAGCATCAGCCC (SEQ ID NO: 13)
The underline designates the self-annealing (complementary) sequences.
Component Volume (111) for 25 ul reaction
4G2 VL amplification primers (10 uM each) 0.5
4G2 RNA 0.1 (20 ng)
2X OneTaq One-Step RT-PCR Buffer (NEB) 12.5
OneTaq One-Step RT-PCR enzyme (NEB) 1
Oligo-conjugated beads 2
Mismatch_VL_Fwd oligo (varying concentrations, as listed below) 2.5
Water 4
Optiprep 2
RNase inhibitor 0.5
4G2 VL Amplification Fwd Primer Rev Primer
Primer Mix
# 1 _Fwd_Non-SA Mus_Kappa_ReV_Biotin
# 2 4G2_VL_Fwd_SA Mus_Kappa_ReV_Biotin
For competing mismatch oligo concentration used with the forward self-annealing primer
(4G2_VL_Fwd_SA), the following conditions were used:
Molar Ratio of Fwd Oligos ch_VL_Fwd_SA
(Mismatch_VL_Fwd_SA : concentration (11M)
4G2_VL_Fwd_SA)
:1 30
:1 10
:1 5
1.25:1 2.5
0.625: 1 1.25
0:1 0
For competing mismatch oligo concentration used with the Fwd lf—annealing primer
(4G2_VL_Fwd_Non-SA), the following conditions were used:
Molar Ratio of Fwd Oligos
Mismatch_VL_Fwd_Non-
(Mismatch_VL_Fwd_Non-SA :
SA concentration (11M)
4G2_VL_Fwd_Non-SA )
:1 10
1:1 2
0.1:1 0.2
0.01:1 0.02
0:1 0
The samples were thermocycled with the following program:
After cycling, the samples were gently mixed by pipetting to suspend the beads. The
samples were then placed in a thermocycler with the following program to facilitate PCR product
capture onto beads:
Step Temp Time
1 70 0C 30 sec
2 55 OC 4 min
3 50 OC 4 min
4 45 °C 4 min
40 OC 3 min
7 4 OC hold
After the ing procedure, the tubes were applied to magnets and the supernatants were
recovered. The supernatants were ed by agarose gel electrophoresis to confirm successful
generation of PCR product. The beads, having captured PCR product, were washed three times with
100 pl ice-cold PBSA (1X PBS containing 1% BSA) using a magnet to sequester beads. Each sample
was then incubated with 100 pl of 1:500 AlexaFluor 647 IgG Fraction Monoclonal Mouse Anti-Biotin
(Jackson) in PBSA with on at 4°C and away from light for 45 minutes. After incubation, the
beads were washed three times with 100 pl ice-cold PBSA. After a final wash, beads were
resuspended with 100 pl ice-cold PBSA and analyzed with a flow cytometer for AlexaGluo647
fluorescence signal.
Mean fluorescence intensity (MFI) was determined for each sample. The sample data were
plotted as a percentage of MFI ve to MFI corresponding to absence of mismatch oligo. As
shown in FIGS. 7A-7B, only the self-annealing primers were capable of preventing PCR product
capture competition at high ratios of mismatched primer oligo to d primer oligo. These results
show that, specifically with the self-annealing primer design, the PCR product can be efficiently
captured under conditions of d more unused primer — conditions that, for example, simulate
multiple antibody repertoire conditions.
INCORPORATION BY REFERENCE
All ations, patents, and Accession numbers mentioned herein are hereby incorporated
by reference in their ty as if each individual publication or patent was specifically and
individually indicated to be incorporated by reference.
LENTS
While specific embodiments of the subject invention have been discussed, the above
specification is illustrative and not restrictive. Many variations of the ion will become apparent
to those skilled in the art upon review of this specification and the claims below. The full scope of the
invention should be determined by reference to the claims, along with their full scope of equivalents,
and the specification, along with such variations.
2017/068204
Claims (47)
1. A method of making a c acid sequence comprising a sequence that encodes a heavy chain element (HC element) of an antibody heavy chain variable region (HCVR) and a light chain element (LC element) of an dy light chain variable region (LCVR), and wherein the HCVR and LCVR are matched, the method comprising: a) acquiring an isolated production reaction site, sing: i) a heavy chain (HC) strand, n the HC strand is a strand of a heavy chain double-stranded cDNA (HC ds cDNA) comprising a segment that encodes the HC element of 10 the HCVR from a cell; and ii) a light chain (LC) , wherein the LC strand is a strand of a light chain double- stranded cDNA (LC ds cDNA) comprising a segment that encodes the LC element of the LCVR from the cell, and b) covalent linking of the HC strand to the LC strand, 15 n the isolated production reaction site does not comprise a nucleic acid encoding an HCVR or an LCVR from a cell other than the cell, thereby making the nucleic acid sequence.
2. The method of claim 1, wherein the HC element ses, or consists of, a heavy chain 20 variable region sequence (HCVRS), or an antigen binding fragment thereof.
3. The method of claim 1 or 2, wherein the LC element comprises, or consists of, a light chain variable region sequence (LCVRS), or an antigen binding fragment thereof. 25
4. The method of any of claims 1-3, wherein the nucleic acid sequence is configured such that, when expressed, the HC t and the LC element form a functional antigen g molecule in vilro, ex vivo, or in vivo.
5. The method of any of claims 1-4, n acquiring the isolated production reaction site 30 comprises: a) acquiring a capture substrate bound to: (i) a first double-stranded cDNA (ds cDNA) comprising a strand that is complementary to a first mRNA that encodes the HCVR from the cell; and (ii) a second ds cDNA comprising a strand complementary to a second mRNA 35 encoding the LCVR from the cell, to produce a loaded capture substrate, and b) maintaining the isolated production reaction site under conditions that allow amplification of the first and second ds cDNAs, to produce: a plurality of HC ds cDNAs comprising the segment that encodes the HC element of the HCVR from the cell; and a plurality of LC ds cDNAs comprising the segment that encodes an LC element of the LCVR from the cell.
6. The method of any of claims 1-5, wherein the capture substrate comprises a bead and a moiety which binds to cDNA.
7. The method of any of claims 1-6, wherein the isolated production on site comprises a 10 reagent mixture suitable for producing, from the first and second mRNAs, a first ds cDNA comprising the t that encodes the HC element of the HCVR of the cell, and a second ds cDNA comprising a segment that encodes the LC element of the LCVR of the cell.
8. The method of any of claims 5-7, wherein the first and second ds cDNAs are amplified in 15 the ce of primers, wherein at least one of the s comprises a first member, a second member, and a nucleotide modification between the first and second members, wherein the nucleotide modification reduces DNA synthesis.
9. The method of claim 8, wherein the nucleotide modification comprises an insertion of a 20 spacer between two adjacent nucleotides or a modification to a ribose.
10. The method of claim 8 or 9, wherein the first member is capable of annealing with the second member in the same primer or a different primer, forming a double-stranded structure comprising a duplex region of 4, 5,6, 7, 8, 9, 10, ll, 12, 13, 14, 15, 16, 17, 18, 19, 20, more 25 basepairs.
11. The method of any of claims 8-10, n at least one of the primers is phosphorylated and comprises a sequence ng at least a portion of a linker sequence, or a complementary sequence thereof.
12. The method of any of claims 1-1 1, wherein the HC ds cDNA comprises a 5’ overhang and a blunt end and the LC ds cDNA comprises a 5’ overhang and a blunt end.
13. The method of any of claims 1-12, wherein the HC strand and the LC strand are 35 covalently linked to produce a single stranded c acid sequence, wherein the HC and LC strands are both sense s or both antisense strands.
14. The method of any of claims 1-13, wherein the covalent linking occurs in an isolated linkage reaction site comprising a ligase.
15. The method of any of claims 1-14, wherein the HC strand and the LC strand are covalently linked in the presence of a splint oligonucleotide, wherein the splint oligonucleotide is hybridized to a sequence comprising the junction of the HC strand and the LC strand to form a duplexed region at the site of linkage.
16. The method of claim 15, wherein the splint oligonucleotide comprises a modification that 10 inhibits DNA synthesis.
17. The method of any of claims l-l6, further comprising, prior to acquiring the isolated production reaction site, acquiring an mRNA loaded capture substrate comprising: a) acquiring an isolated cell reaction site, comprising: 15 i) a cell; and ii) a e substrate capable of binding a first mRNA encoding an HCVR from the cell and a second mRNA encoding an LCVR from the cell; and b) maintaining the isolated cell reaction site under conditions that allow lysis of the cell and binding of the capture substrate with the first mRNA and the second mRNA to form the mRNA 20 loaded capture substrate, wherein the isolated cell on site does not include a nucleic acid encoding an HCVR or an LCVR from a cell other than the cell.
18. The method of claim 17, further comprising releasing the mRNA loaded e substrate 25 from the ed cell reaction site in the ce of a poly(dA) or poly(dT) oligonucleotide.
19. The method of any of claims 1-18, further comprising ying the nucleic acid sequence. 30
20. The method of any of claims 1-19, further comprising sequencing all or a portion of the nucleic acid sequence.
21. The method of any of claims 1-20, r comprising inserting all or a portion of nucleic acid ce into a vector.
22. The method of any of claims 1-21, comprising expressing the nucleic acid sequence to produce a polypeptide comprising the segment that encodes the HC element of the HCVR, and the segment that encodes the LC element of the LCVR.
23. The method of claim 22, further comprising contacting the polypeptide with an antigen and determining if the polypeptide binds the n.
24. A method of making a library comprising a plurality of unique members, the method comprising: 10 making the plurality of members by the method of any of claims 1-23, wherein each of the members comprises a sequence that encodes a heavy chain element (HC element) of a heavy chain variable region (HCVR) and a light chain element (LC element) of a light chain variable region (LCVR), n the HCVR and the LCVR are matched, and wherein each unique nucleic acid sequence of the ity comprises an HC element and an LC element from a 15 different unique cell, thereby making the library.
25. The method of claim 24, wherein the y comprises one, two, three, or all of the following properties: 20 a) the plurality of unique members ses at least 104, 105, 106, 107, 108, or 109 unique members; b) the plurality of unique members comprises 104 to 109, 104 to 108, 104 to 107, 104 to 106, 104 to 105, 108 to 109, 107 to 109, 106 to 109, 105 to 109, 105 to 103, 106 to 107, 104 to 105, 105 to 106, 106 to 107, 107 to 108, or 108 to 109 unique members; 25 c) at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%, of the members in the y are unique members,; or d) less than 20%, 15%, 10%, 5%, 4%, 3%, 2%, or 1%, of the members in the library are unique members. 30
26. A library made by the method of claim 25.
27. The library of claim 26, wherein the library is a display library.
28. A method of making a g polypeptide, the method comprising: 35 a) acquiring a y of claim 26 or 27; and b) expressing a polypeptide encoded by a unique nucleic acid of the library.
29. The method of claim 28, further sing ting the polypeptide with an n and obtaining a nucleic acid that encodes a polypeptide that binds the n.
30. A method of making a nucleic acid sequence comprising a sequence that encodes an or chain element (AC element) of a T-cell receptor (TCR) or chain variable region (ACVR) and a [5 chain element (BC element) of a TCR [3 chain variable region (BCVR), and wherein the ACVR and BCVR are matched, the method comprising: a) acquiring an isolated production reaction site comprising: i) an or chain (AC) strand, wherein the AC strand is a strand of an a chain double- 10 stranded cDNA (AC ds cDNA) comprising a segment that encodes an AC element of the ACVR from a cell; and ii) a [3 chain (BC) strand, wherein the BC strand is a strand of a [3 chain - stranded cDNA (BC ds cDNA) comprising a segment that encodes a BC element of the BCVR from the cell, and 15 b) covalent linking of an AC strand to a BC strand, wherein the isolated production reaction site does not comprise a nucleic acid encoding a BCVR or an ACVR from a cell other than the cell, thereby making the nucleic acid sequence. 20
31. The method of claim 30, wherein the nucleic acid sequence is configured such that, when expressed, the AC element and the BC element form a functional antigen binding molecule.
32. The method of claim 31, wherein the covalent linking occurs in an isolated linkage reaction site comprising a ligase.
33. A method of making a library comprising a plurality of unique members, the method comprising: making the ity of members by the method of any of claims 30-32, wherein each of the members comprises a sequence that encodes a or chain element (AC 30 element) of a or chain variable region (ACVR) and a [3 chain element (BC element) of a [3 chain le region , wherein the ACVR and BCVR are d, and wherein each unique nucleic acid sequence of the plurality comprises an AC element and a BC element from a different unique cell, thereby making the library.
34. A library made by the method of claim 33.
35. A method of making a g polypeptide, the method comprising: a) acquiring the library of claim 34; and b) expressing a polypeptide encoded by a unique nucleic acid of the library.
36. A method of making a nucleic acid sequence sing a ce that encodes an 7 chain element (GC element) of a TCR 7 chain variable region (GCVR) and a 5 chain element (DC element) of a TCR 5 chain variable region (DCVR), and wherein the GCVR and DCVR are matched, the method comprising: a) acquiring an isolated production reaction site, comprising: 10 i) a y chain (GC) strand, wherein the GC strand is a strand of a 7 chain double- stranded cDNA (GC ds cDNA) sing a segment that encodes a GC element of the GCVR from a cell; and ii) a 5 chain (DC) , wherein the DC strand is a strand of a 5 chain double- stranded cDNA (DC ds cDNA) comprising a segment that encodes a DC element of the 15 DCVR from the cell, and b) covalent linking of a GC strand to a DC strand, wherein the isolated production reaction site does not comprises a nucleic acid ng a DCVR or a GCVR from a cell other than the cell, thereby making the c acid sequence.
37. The method of claim 36, wherein the nucleic acid sequence is configured such that, when expressed, the GC t and the DC element form a functional antigen binding molecule.
38. The method of claim 36 or 37, wherein the covalent linking occurs in an isolated linkage 25 reaction site comprising a ligase.
39. A method of making a library comprising a plurality of unique members, the method comprising: making the plurality of members by the method of any of claims 36-38, 30 wherein each of the members ses a sequence that encodes a 7 chain element (GC element) of a 1/ chain variable region (GCVR) and a 5 chain element (DC element) of a 5 chain variable region (DCVR), wherein the GCVR and DCVR are matched, and wherein each unique nucleic acid sequence of the plurality ses a GC element and a DC element from a different unique cell, 35 thereby making the library.
40. A library made by the method of claim 39.
41. A method of making a g polypeptide, the method comprising: a) acquiring the library of claim 40; and b) expressing a polypeptide encoded by a unique nucleic acid of the library.
42. An isolated production reaction site, sing: a) a heavy chain (HC) strand, wherein the HC strand is a strand of a heavy chain double- stranded cDNA (HC ds cDNA) comprising a segment that encodes an HC element of the HCVR from a cell; and 10 b) a light chain (LC) strand, wherein the LC strand is a strand of a light chain double-stranded cDNA (LC ds cDNA) comprising a segment that encodes an LC element of the LCVR from the cell, c) a primer comprising a first member, a second member, and a nucleotide ation between the first and second members, n the nucleotide modification s DNA synthesis, wherein the HCVR and LCVR are matched, and wherein the ed production reaction site 15 does not comprise a nucleic acid encoding an LCVR or an HCVR from a cell other than the cell.
43. The isolated production reaction site of claim 42, wherein the first member is capable of annealing with the second member in the same primer or a different primer, forming double-stranded structure comprising a duplex region of 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, more basepairs. .
44. A self-annealing oligonucleotide comprising a first member, a second member, and a spacer located between the first and second members, wherein the first member is capable of annealing with the second member in the same oligonucleotide or a different ucleotide, forming 25 a hairpin structure or a -stranded structure comprising a duplex region of 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19,20, more irs.
45. The oligonucleotide of claim 44, wherein the spacer is a flexible spacer or a PEG spacer. 30
46. An isolated linkage reaction site, comprising: a) a heavy chain (HC) strand, wherein the HC strand is a strand of a heavy chain double- stranded cDNA (HC ds cDNA) comprising a segment that encodes an HC element of the HCVR from a cell; b) a light chain (LC) strand, wherein the LC strand is a strand of a light chain double-stranded 35 cDNA (LC ds cDNA) sing a segment that encodes an LC element of the LCVR from the cell; c) a splint oligonucleotide that is capable of hybridizing to a ce comprising the junction of the HC strand and the LC strand, to form a duplexed region at the site of linkage, wherein the HCVR and LCVR are matched, wherein the HC strand and the LC strand are covalently linked, and wherein the ed linkage reaction site does not comprise a nucleic acid encoding an HCVR or an LCVR from a cell other than the cell,
47. The isolated linkage reaction site of claim 46, further comprising a ligase.
Applications Claiming Priority (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US62/438,712 | 2016-12-23 |
Publications (1)
Publication Number | Publication Date |
---|---|
NZ753978A true NZ753978A (en) |
Family
ID=
Similar Documents
Publication | Publication Date | Title |
---|---|---|
TWI829621B (en) | Anti-human vista antibodies and use thereof | |
TWI828625B (en) | Guidance and navigation control proteins and method of making and using thereof | |
JP6731403B2 (en) | Compositions and methods for use in enhancing immune response and cancer treatment | |
AU2015357463B2 (en) | Identification of VSIG8 as the putative vista receptor and its use thereof to produce vista/VSIG8 modulators | |
JP7399852B2 (en) | Multispecific antibodies and their production and use methods | |
CN117285641A (en) | Guidance and navigation control proteins and methods of making and using the same | |
US20240141334A1 (en) | Binding polypeptides and methods of making the same | |
JP2020530306A (en) | Multispecific antibody and its preparation and usage | |
WO2013190555A1 (en) | Lsr antibodies, and uses thereof for treatment of cancer | |
AU2017341936A1 (en) | Modular tetravalent bispecific antibody platform | |
WO2016089610A1 (en) | Bispecific antibody for cancer immunotherapy | |
NZ753978A (en) | Binding polypeptides and methods of making the same | |
NZ795976A (en) | Binding polypeptides and methods of making the same | |
US20220185911A1 (en) | Therapeutic antibodies for treating lung cancer | |
TW201625697A (en) | Compositions and methods of use for augmented immune response, and cancer therapy | |
TW202328196A (en) | Treatment and prevention of cancer using vista antigen-binding molecules | |
IL256261A (en) | Hide1 compositions and methods |